Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Nature ; 609(7926): 335-340, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35853476

RESUMO

Adhesive pili assembled through the chaperone-usher pathway are hair-like appendages that mediate host tissue colonization and biofilm formation of Gram-negative bacteria1-3. Archaic chaperone-usher pathway pili, the most diverse and widespread chaperone-usher pathway adhesins, are promising vaccine and drug targets owing to their prevalence in the most troublesome multidrug-resistant pathogens1,4,5. However, their architecture and assembly-secretion process remain unknown. Here, we present the cryo-electron microscopy structure of the prototypical archaic Csu pilus that mediates biofilm formation of Acinetobacter baumannii-a notorious multidrug-resistant nosocomial pathogen. In contrast to the thick helical tubes of the classical type 1 and P pili, archaic pili assemble into an ultrathin zigzag architecture secured by an elegant clinch mechanism. The molecular clinch provides the pilus with high mechanical stability as well as superelasticity, a property observed for the first time, to our knowledge, in biomolecules, while enabling a more economical and faster pilus production. Furthermore, we demonstrate that clinch formation at the cell surface drives pilus secretion through the outer membrane. These findings suggest that clinch-formation inhibitors might represent a new strategy to fight multidrug-resistant bacterial infections.


Assuntos
Acinetobacter baumannii , Microscopia Crioeletrônica , Fímbrias Bacterianas , Chaperonas Moleculares , Acinetobacter baumannii/citologia , Acinetobacter baumannii/ultraestrutura , Elasticidade , Proteínas de Fímbrias/química , Proteínas de Fímbrias/metabolismo , Proteínas de Fímbrias/ultraestrutura , Fímbrias Bacterianas/química , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/ultraestrutura , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/ultraestrutura
2.
Nat Commun ; 13(1): 749, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35136069

RESUMO

Tousled-like kinases (TLKs) are nuclear serine-threonine kinases essential for genome maintenance and proper cell division in animals and plants. A major function of TLKs is to phosphorylate the histone chaperone proteins ASF1a and ASF1b to facilitate DNA replication-coupled nucleosome assembly, but how TLKs selectively target these critical substrates is unknown. Here, we show that TLK2 selectivity towards ASF1 substrates is achieved in two ways. First, the TLK2 catalytic domain recognizes consensus phosphorylation site motifs in the ASF1 C-terminal tail. Second, a short sequence at the TLK2 N-terminus docks onto the ASF1a globular N-terminal domain in a manner that mimics its histone H3 client. Disrupting either catalytic or non-catalytic interactions through mutagenesis hampers ASF1 phosphorylation by TLK2 and cell growth. Our results suggest that the stringent selectivity of TLKs for ASF1 is enforced by an unusual interaction mode involving mutual recognition of a short sequence motifs by both kinase and substrate.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Mimetismo Molecular , Proteínas Quinases/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Domínio Catalítico/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/isolamento & purificação , Proteínas de Ciclo Celular/ultraestrutura , Sequência Conservada , Cristalografia por Raios X , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/isolamento & purificação , Chaperonas Moleculares/ultraestrutura , Simulação de Acoplamento Molecular , Mutagênese , Biblioteca de Peptídeos , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/isolamento & purificação , Proteínas Quinases/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Especificidade por Substrato
3.
Nature ; 601(7893): 465-469, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34937936

RESUMO

Hsp90 is a conserved and essential molecular chaperone responsible for the folding and activation of hundreds of 'client' proteins1-3. The glucocorticoid receptor (GR) is a model client that constantly depends on Hsp90 for activity4-9. GR ligand binding was previously shown to nr inhibited by Hsp70 and restored by Hsp90, aided by the co-chaperone p2310. However, a molecular understanding of the chaperone-mediated remodelling that occurs between the inactive Hsp70-Hsp90 'client-loading complex' and an activated Hsp90-p23 'client-maturation complex' is lacking for any client, including GR. Here we present a cryo-electron microscopy (cryo-EM) structure of the human GR-maturation complex (GR-Hsp90-p23), revealing that the GR ligand-binding domain is restored to a folded, ligand-bound conformation, while being simultaneously threaded through the Hsp90 lumen. In addition, p23 directly stabilizes native GR using a C-terminal helix, resulting in enhanced ligand binding. This structure of a client bound to Hsp90 in a native conformation contrasts sharply with the unfolded kinase-Hsp90 structure11. Thus, aided by direct co-chaperone-client interactions, Hsp90 can directly dictate client-specific folding outcomes. Together with the GR-loading complex structure12, we present the molecular mechanism of chaperone-mediated GR remodelling, establishing the first, to our knowledge, complete chaperone cycle for any Hsp90 client.


Assuntos
Microscopia Crioeletrônica , Proteínas de Choque Térmico HSP90 , Prostaglandina-E Sintases , Receptores de Glucocorticoides , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP70/ultraestrutura , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP90/ultraestrutura , Humanos , Ligantes , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/ultraestrutura , Prostaglandina-E Sintases/química , Prostaglandina-E Sintases/metabolismo , Prostaglandina-E Sintases/ultraestrutura , Ligação Proteica , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/ultraestrutura
4.
Sci Rep ; 11(1): 12515, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131228

RESUMO

The glucocorticoid receptor is a key regulator of essential physiological processes, which under the control of the Hsp90 chaperone machinery, binds to steroid hormones and steroid-like molecules and in a rather complicated and elusive response, regulates a set of glucocorticoid responsive genes. We here examine a human glucocorticoid receptor variant, harboring a point mutation in the last C-terminal residues, L773P, that was associated to Primary Generalized Glucocorticoid Resistance, a condition originating from decreased affinity to hormone, impairing one or multiple aspects of GR action. Using in vitro and in silico methods, we assign the conformational consequences of this mutation to particular GR elements and report on the altered receptor properties regarding its binding to dexamethasone, a NCOA-2 coactivator-derived peptide, DNA, and importantly, its interaction with the chaperone machinery of Hsp90.


Assuntos
Glucocorticoides/genética , Proteínas de Choque Térmico HSP90/genética , Conformação Molecular/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Animais , DNA/genética , Dexametasona/farmacologia , Glucocorticoides/química , Proteínas de Choque Térmico HSP90/ultraestrutura , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/patologia , Chaperonas Moleculares/genética , Chaperonas Moleculares/ultraestrutura , Coativador 2 de Receptor Nuclear/química , Coativador 2 de Receptor Nuclear/genética , Peptídeos/genética , Mutação Puntual/genética , Ligação Proteica/genética , Receptores de Glucocorticoides/deficiência , Receptores de Glucocorticoides/ultraestrutura
5.
Nat Commun ; 12(1): 3174, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34039964

RESUMO

Chaperones Tapasin and TAP-binding protein related (TAPBPR) perform the important functions of stabilizing nascent MHC-I molecules (chaperoning) and selecting high-affinity peptides in the MHC-I groove (editing). While X-ray and cryo-EM snapshots of MHC-I in complex with TAPBPR and Tapasin, respectively, have provided important insights into the peptide-deficient MHC-I groove structure, the molecular mechanism through which these chaperones influence the selection of specific amino acid sequences remains incompletely characterized. Based on structural and functional data, a loop sequence of variable lengths has been proposed to stabilize empty MHC-I molecules through direct interactions with the floor of the groove. Using deep mutagenesis on two complementary expression systems, we find that important residues for the Tapasin/TAPBPR chaperoning activity are located on a large scaffolding surface, excluding the loop. Conversely, loop mutations influence TAPBPR interactions with properly conformed MHC-I molecules, relevant for peptide editing. Detailed biophysical characterization by solution NMR, ITC and FP-based assays shows that the loop hovers above the MHC-I groove to promote the capture of incoming peptides. Our results suggest that the longer loop of TAPBPR lowers the affinity requirements for peptide selection to facilitate peptide loading under conditions and subcellular compartments of reduced ligand concentration, and to prevent disassembly of high-affinity peptide-MHC-I complexes that are transiently interrogated by TAPBPR during editing.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulinas/metabolismo , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Antígenos/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , Técnicas de Inativação de Genes , Células HEK293 , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/isolamento & purificação , Antígenos de Histocompatibilidade Classe I/ultraestrutura , Humanos , Imunoglobulinas/genética , Imunoglobulinas/isolamento & purificação , Imunoglobulinas/ultraestrutura , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Proteínas de Membrana/ultraestrutura , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/ultraestrutura , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Biblioteca de Peptídeos , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura
6.
FEBS J ; 288(7): 2222-2237, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33058391

RESUMO

The formation of ordered Z (Glu342Lys) α1 -antitrypsin polymers in hepatocytes is central to liver disease in α1 -antitrypsin deficiency. In vitro experiments have identified an intermediate conformational state (M*) that precedes polymer formation, but this has yet to be identified in vivo. Moreover, the mechanism of polymer formation and their fate in cells have been incompletely characterised. We have used cell models of disease in conjunction with conformation-selective monoclonal antibodies and a small molecule inhibitor of polymerisation to define the dynamics of polymer formation, accumulation and secretion. Pulse-chase experiments demonstrate that Z α1 -antitrypsin accumulates as short-chain polymers that partition with soluble cellular components and are partially secreted by cells. These precede the formation of larger, insoluble polymers with a longer half-life (10.9 ± 1.7 h and 20.9 ± 7.4 h for soluble and insoluble polymers, respectively). The M* intermediate (or a by-product thereof) was identified in the cells by a conformation-specific monoclonal antibody. This was completely abrogated by treatment with the small molecule, which also blocked the formation of intracellular polymers. These data allow us to conclude that the M* conformation is central to polymerisation of Z α1 -antitrypsin in vivo; preventing its accumulation represents a tractable approach for pharmacological treatment of this condition; polymers are partially secreted; and polymers exist as two distinct populations in cells whose different dynamics have likely consequences for the aetiology of the disease.


Assuntos
Chaperonas Moleculares/genética , Conformação Proteica/efeitos dos fármacos , Deficiência de alfa 1-Antitripsina/tratamento farmacológico , alfa 1-Antitripsina/genética , Anticorpos Monoclonais/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/química , Chaperonas Moleculares/ultraestrutura , Polímeros/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , alfa 1-Antitripsina/química , alfa 1-Antitripsina/efeitos dos fármacos , alfa 1-Antitripsina/ultraestrutura , Deficiência de alfa 1-Antitripsina/genética
7.
Biochim Biophys Acta Proteins Proteom ; 1869(2): 140576, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33253897

RESUMO

HdeA is a small acid-stress chaperone protein with a unique activity profile. At physiological pH, it forms a folded, but inactive, dimer. Below pH 3.0, HdeA unfolds and dissociates into disordered monomers, utilizing exposed hydrophobic patches to bind other unfolded proteins and prevent their irreversible aggregation. In this way, HdeA has a key role in helping pathogenic bacteria survive our acidic stomach and colonize our intestines, facilitating the spread of dysentery. Despite numerous publications on the topic, there remain questions about the mechanism by which HdeA unfolding and activation are triggered. Previous studies usually assessed HdeA unfolding over pH increments that are too far apart to gain fine detail of the process of unfolding and dimer dissociation, and often employed techniques that prevented thorough evaluation of specific regions of the protein. We used a variety of heteronuclear NMR experiments to investigate changes to backbone and side chain structure and dynamics of HdeA at four pHs between 3.0 and 2.0. We found that the long loop in the dimer interface is an early site of initiation of dimer dissociation, and that a molecular "clasp" near the disulfide bond is broken open at low pH as part, or as a trigger, of unfolding; this process also results in the separation of C-terminal helices and exposure of key hydrophobic client binding sites. Our results highlight important regions of HdeA that may have previously been overlooked because they lie too close to the disulfide bond or are thought to be too dynamic in the folded state to influence unfolding processes.


Assuntos
Proteínas de Escherichia coli/genética , Escherichia coli/genética , Chaperonas Moleculares/genética , Conformação Proteica , Ácidos/farmacologia , Escherichia coli/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Humanos , Concentração de Íons de Hidrogênio , Chaperonas Moleculares/química , Chaperonas Moleculares/ultraestrutura , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Multimerização Proteica/genética , Desdobramento de Proteína
8.
FEBS J ; 288(9): 2870-2883, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-32979284

RESUMO

Proteins destined to various intra- and extra-cellular locations must traverse membranes most frequently in an unfolded form. When the proteins being translocated need to remain in a folded state, specialized cellular transport machinery is used. One such machine is the membrane-bound AAA protein Bcs1 (Bcs1), which assists the iron-sulfur protein, an essential subunit of the respiratory Complex III, across the mitochondrial inner membrane. Recent structure determinations of mouse and yeast Bcs1 in three different nucleotide states reveal its homo-heptameric association and at least two dramatically different conformations. The apo and ADP-bound structures are similar, both containing a large substrate-binding cavity accessible to the mitochondrial matrix space, which contracts by concerted motion of the ATPase domains upon ATP binding, suggesting that bound substrate could then be pushed across the membrane. ATP hydrolysis drives substrate release and resets Bcs1 conformation back to the apo/ADP form. These structures shed new light on the mechanism of folded protein translocation across a membrane, provide better understanding on the assembly process of the respiratory Complex III, and correlate clinical presentations of disease-associated mutations with their locations in the 3D structure.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Chaperonas Moleculares/genética , Conformação Proteica , Proteínas de Saccharomyces cerevisiae/genética , ATPases Associadas a Diversas Atividades Celulares/ultraestrutura , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/genética , Animais , Complexo III da Cadeia de Transporte de Elétrons/genética , Complexo III da Cadeia de Transporte de Elétrons/ultraestrutura , Humanos , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/ultraestrutura , Camundongos , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/ultraestrutura , Chaperonas Moleculares/ultraestrutura , Domínios Proteicos/genética , Dobramento de Proteína , Transporte Proteico/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestrutura
9.
IET Nanobiotechnol ; 14(6): 491-500, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32755959

RESUMO

Manipulating molecular scale bio-nanorobots and influencing their behaviour is one of the major challenges of new researches. Many coiled coil type proteins are involved in important biological functions due to physical properties that make them ideal for both nanoscale manipulation and sensing. The Prefoldin beta subunit from Thermococcus strain KS-1(Prefoldin ß1) is one of the possible proteins that can serve as a new bio-nano-actuator. Besides having a balanced architecture, Prefoldin ß1 can exhibit a wide range of exclusive authorities. In this study, steered molecular dynamics simulation is applied along with the centre of mass pulling and analyses of Prefoldin ß1 conformational changes to characterise some of those abilities. Thus, applying external mechanical force without any position constraint shows that it has no movement throughout simulations. This proposes a novel method to capture different sizes and shapes of cargoes. During simulations, each arm was found to be very flexible, allowing it to enlarge its central cavity and capture different cargoes. For a more accurate analysis, the variations in the cavity of nano-actuator are investigated qualitatively and quantitatively with different parameters. Also, the force analysis of the arms can provide us with decent information about the performance of this nano-actuator.


Assuntos
Chaperonas Moleculares , Fenômenos Biomecânicos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/ultraestrutura , Simulação de Dinâmica Molecular , Nanoestruturas/química , Nanoestruturas/ultraestrutura , Conformação Proteica , Thermococcus/química
10.
J Mol Biol ; 432(14): 4127-4138, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32450081

RESUMO

The multi-domain RNA binding protein RBM5 is a molecular signature of metastasis. RBM5 regulates alternative splicing of apoptotic genes including the cell death receptor Fas and the initiator Caspase-2. The RBM5 RanBP2-type zinc finger (Zf1) is known to specifically recognize single-stranded RNAs with high affinity. Here, we study the structure and conformational dynamics of the Zf1 zinc finger of human RBM5 using NMR. We show that the presence of a non-canonical cysteine in Zf1 kinetically destabilizes the protein. Metal-exchange kinetics show that mutation of the cysteine establishes high-affinity coordination of the zinc. Our data indicate that selection of such a structurally destabilizing mutation during the course of evolution could present an opportunity for functional adaptation of the protein.


Assuntos
Proteínas de Ciclo Celular/ultraestrutura , Proteínas de Ligação a DNA/ultraestrutura , Chaperonas Moleculares/ultraestrutura , Complexo de Proteínas Formadoras de Poros Nucleares/ultraestrutura , Proteínas de Ligação a RNA/ultraestrutura , Proteínas Supressoras de Tumor/ultraestrutura , Dedos de Zinco/genética , Processamento Alternativo/genética , Sequência de Aminoácidos/genética , Apoptose/genética , Caspase 2/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Ligação Proteica/genética , Conformação Proteica , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Zinco/química
11.
Cell Rep ; 30(11): 3699-3709.e6, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32126208

RESUMO

Many chaperones promote nascent polypeptide folding followed by substrate release through ATP-dependent conformational changes. Here we show cryoEM structures of Gα subunit folding intermediates in complex with full-length Ric-8A, a unique chaperone-client system in which substrate release is facilitated by guanine nucleotide binding to the client G protein. The structures of Ric-8A-Gαi and Ric-8A-Gαq complexes reveal that the chaperone employs its extended C-terminal region to cradle the Ras-like domain of Gα, positioning the Ras core in contact with the Ric-8A core while engaging its switch2 nucleotide binding region. The C-terminal α5 helix of Gα is held away from the Ras-like domain through Ric-8A core domain interactions, which critically depend on recognition of the Gα C terminus by the chaperone. The structures, complemented with biochemical and cellular chaperoning data, support a folding quality control mechanism that ensures proper formation of the C-terminal α5 helix before allowing GTP-gated release of Gα from Ric-8A.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP/química , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Sequência de Aminoácidos , Subunidades alfa de Proteínas de Ligação ao GTP/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/ultraestrutura , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Modelos Biológicos , Modelos Moleculares , Chaperonas Moleculares/ultraestrutura , Fosforilação , Ligação Proteica , Dobramento de Proteína , Estabilidade Proteica , Estrutura Secundária de Proteína , Controle de Qualidade
12.
Proc Natl Acad Sci U S A ; 117(14): 7814-7823, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32198203

RESUMO

Hsp70 is a conserved molecular chaperone that plays an indispensable role in regulating protein folding, translocation, and degradation. The conformational dynamics of Hsp70 and its regulation by cochaperones are vital to its function. Using bulk and single-molecule fluorescence resonance energy transfer (smFRET) techniques, we studied the interdomain conformational distribution of human stress-inducible Hsp70A1 and the kinetics of conformational changes induced by nucleotide and the Hsp40 cochaperone Hdj1. We found that the conformations between and within the nucleotide- and substrate-binding domains show heterogeneity. The conformational distribution in the ATP-bound state can be induced by Hdj1 to form an "ADP-like" undocked conformation, which is an ATPase-stimulated state. Kinetic measurements indicate that Hdj1 binds to monomeric Hsp70 as the first step, then induces undocking of the two domains and closing of the substrate-binding cleft. Dimeric Hdj1 then facilitates dimerization of Hsp70 and formation of a heterotetrameric Hsp70-Hsp40 complex. Our results provide a kinetic view of the conformational cycle of Hsp70 and reveal the importance of the dynamic nature of Hsp70 for its function.


Assuntos
Proteínas de Choque Térmico HSP40/genética , Proteínas de Choque Térmico HSP70/ultraestrutura , Chaperonas Moleculares/ultraestrutura , Conformação Proteica , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/química , Transferência Ressonante de Energia de Fluorescência , Heterogeneidade Genética , Proteínas de Choque Térmico HSP40/química , Proteínas de Choque Térmico HSP70/química , Humanos , Cinética , Modelos Moleculares , Chaperonas Moleculares/química , Ligação Proteica/genética , Domínios Proteicos/genética , Dobramento de Proteína , Multimerização Proteica/genética
13.
Nucleic Acids Res ; 48(3): 1531-1550, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31807785

RESUMO

FKBP53 is one of the seven multi-domain FK506-binding proteins present in Arabidopsis thaliana, and it is known to get targeted to the nucleus. It has a conserved PPIase domain at the C-terminus and a highly charged N-terminal stretch, which has been reported to bind to histone H3 and perform the function of a histone chaperone. To better understand the molecular details of this PPIase with histone chaperoning activity, we have solved the crystal structures of its terminal domains and functionally characterized them. The C-terminal domain showed strong PPIase activity, no role in histone chaperoning and revealed a monomeric five-beta palm-like fold that wrapped over a helix, typical of an FK506-binding domain. The N-terminal domain had a pentameric nucleoplasmin-fold; making this the first report of a plant nucleoplasmin structure. Further characterization revealed the N-terminal nucleoplasmin domain to interact with H2A/H2B and H3/H4 histone oligomers, individually, as well as simultaneously, suggesting two different binding sites for H2A/H2B and H3/H4. The pentameric domain assists nucleosome assembly and forms a discrete complex with pre-formed nucleosomes; wherein two pentamers bind to a nucleosome.


Assuntos
Proteínas de Arabidopsis/ultraestrutura , Histonas/genética , Chaperonas Moleculares/ultraestrutura , Nucleoplasminas/química , Proteínas de Ligação a Tacrolimo/ultraestrutura , Arabidopsis/química , Arabidopsis/genética , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Sítios de Ligação/genética , Montagem e Desmontagem da Cromatina/genética , Cristalografia por Raios X , Histonas/química , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Nucleoplasminas/genética , Nucleossomos/química , Nucleossomos/genética , Peptidilprolil Isomerase/genética , Ligação Proteica/genética , Domínios Proteicos/genética , Dobramento de Proteína , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/genética
14.
Proc Natl Acad Sci U S A ; 117(1): 381-387, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31848241

RESUMO

The vast majority of biological carbon dioxide fixation relies on the function of ribulose 1,5-bisphosphate carboxylase/oxygenase (Rubisco). In most cases the enzyme exhibits a tendency to become inhibited by its substrate RuBP and other sugar phosphates. The inhibition is counteracted by diverse molecular chaperones known as Rubisco activases (Rcas). In some chemoautotrophic bacteria, the CbbQO-type Rca Q2O2 repairs inhibited active sites of hexameric form II Rubisco. The 2.2-Å crystal structure of the MoxR AAA+ protein CbbQ2 from Acidithiobacillus ferrooxidans reveals the helix 2 insert (H2I) that is critical for Rca function and forms the axial pore of the CbbQ hexamer. Negative-stain electron microscopy shows that the essential CbbO adaptor protein binds to the conserved, concave side of the CbbQ2 hexamer. Site-directed mutagenesis supports a model in which adenosine 5'-triphosphate (ATP)-powered movements of the H2I are transmitted to CbbO via the concave residue L85. The basal ATPase activity of Q2O2 Rca is repressed but strongly stimulated by inhibited Rubisco. The characterization of multiple variants where this repression is released indicates that binding of inhibited Rubisco to the C-terminal CbbO VWA domain initiates a signal toward the CbbQ active site that is propagated via elements that include the CbbQ α4-ß4 loop, pore loop 1, and the presensor 1-ß hairpin (PS1-ßH). Detailed mechanistic insights into the enzyme repair chaperones of the highly diverse CO2 fixation machinery of Proteobacteria will facilitate their successful implementation in synthetic biology ventures.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Acidithiobacillus/enzimologia , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Chaperonas Moleculares/metabolismo , Ribulose-Bifosfato Carboxilase/metabolismo , ATPases Associadas a Diversas Atividades Celulares/genética , ATPases Associadas a Diversas Atividades Celulares/ultraestrutura , Acidithiobacillus/genética , Acidithiobacillus/ultraestrutura , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/ultraestrutura , Proteínas de Transporte/genética , Proteínas de Transporte/ultraestrutura , Domínio Catalítico/genética , Cristalografia por Raios X , Ativação Enzimática , Ensaios Enzimáticos , Microscopia Eletrônica , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/ultraestrutura , Mutagênese Sítio-Dirigida , Multimerização Proteica , Estrutura Secundária de Proteína , Ribulose-Bifosfato Carboxilase/genética , Ribulose-Bifosfato Carboxilase/ultraestrutura
15.
Nat Commun ; 10(1): 3084, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31300652

RESUMO

Resistance to inhibitors of cholinesterase 8A (Ric8A) is an essential regulator of G protein α-subunits (Gα), acting as a guanine nucleotide exchange factor and a chaperone. We report two crystal structures of Ric8A, one in the apo form and the other in complex with a tagged C-terminal fragment of Gα. These structures reveal two principal domains of Ric8A: an armadillo-fold core and a flexible C-terminal tail. Additionally, they show that the Gα C-terminus binds to a highly-conserved patch on the concave surface of the Ric8A armadillo-domain, with selectivity determinants residing in the Gα sequence. Biochemical analysis shows that the Ric8A C-terminal tail is critical for its stability and function. A model of the Ric8A/Gα complex derived from crosslinking mass spectrometry and molecular dynamics simulations suggests that the Ric8A C-terminal tail helps organize the GTP-binding site of Gα. This study lays the groundwork for understanding Ric8A function at the molecular level.


Assuntos
Proteínas do Domínio Armadillo/ultraestrutura , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/ultraestrutura , Chaperonas Moleculares/ultraestrutura , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Bovinos , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Espalhamento a Baixo Ângulo , Difração de Raios X
16.
Small ; 15(20): e1805558, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30920729

RESUMO

Precisely organized enzyme complexes are often found in nature to support complex metabolic reactions in a highly efficient and specific manner. Scaffolding enzymes on artificial materials has thus gained attention as a promising biomimetic strategy to design biocatalytic systems with enhanced productivity. Herein, a versatile scaffolding platform that can immobilize enzymes on customizable nanofibers is reported. An ultrastable self-assembling filamentous protein, the gamma-prefoldin (γ-PFD), is genetically engineered to display an array of peptide tags, which can specifically and stably bind enzymes containing the counterpart domain through simple in vitro mixing. Successful immobilization of proteins along the filamentous template in tunable density is first verified using fluorescent proteins. Then, two different model enzymes, glucose oxidase and horseradish peroxidase, are used to demonstrate that scaffold attachment could enhance the intrinsic catalytic activity of the immobilized enzymes. Considering the previously reported ability of γ-PFD to bind and stabilize a broad range of proteins, the filament's interaction with the bound enzymes may have created a favorable microenvironment for catalysis. It is envisioned that the strategy described here may provide a generally applicable methodology for the scaffolded assembly of multienzymatic complexes for use in biocatalysis.


Assuntos
Glucose Oxidase/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Chaperonas Moleculares/química , Biocatálise , Enzimas Imobilizadas/metabolismo , Fluorescência , Cinética , Chaperonas Moleculares/ultraestrutura
17.
J Mol Biol ; 430(3): 337-347, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29273204

RESUMO

Antibody Fab fragments have been exploited with significant success to facilitate the structure determination of challenging macromolecules as crystallization chaperones and as molecular fiducial marks for single particle cryo-electron microscopy approaches. However, the inherent flexibility of the "elbow" regions, which link the constant and variable domains of the Fab, can introduce disorder and thus diminish their effectiveness. We have developed a phage display engineering strategy to generate synthetic Fab variants that significantly reduces elbow flexibility, while maintaining their high affinity and stability. This strategy was validated using previously recalcitrant Fab-antigen complexes where introduction of an engineered elbow region enhanced crystallization and diffraction resolution. Furthermore, incorporation of the mutations appears to be generally portable to other synthetic antibodies and may serve as a universal strategy to enhance the success rates of Fabs as structure determination chaperones.


Assuntos
Antígenos/química , Microscopia Crioeletrônica/métodos , Fragmentos Fab das Imunoglobulinas/química , Antígenos/ultraestrutura , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/ultraestrutura , Cristalização/métodos , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/ultraestrutura , Biblioteca de Peptídeos , Conformação Proteica , Engenharia de Proteínas , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestrutura
18.
Nat Commun ; 8(1): 2081, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29234026

RESUMO

Protein misfolding and aggregation is increasingly being recognized as a cause of disease. In Alzheimer's disease the amyloid-ß peptide (Aß) misfolds into neurotoxic oligomers and assembles into amyloid fibrils. The Bri2 protein associated with Familial British and Danish dementias contains a BRICHOS domain, which reduces Aß fibrillization as well as neurotoxicity in vitro and in a Drosophila model, but also rescues proteins from irreversible non-fibrillar aggregation. How these different activities are mediated is not known. Here we show that Bri2 BRICHOS monomers potently prevent neuronal network toxicity of Aß, while dimers strongly suppress Aß fibril formation. The dimers assemble into high-molecular-weight oligomers with an apparent two-fold symmetry, which are efficient inhibitors of non-fibrillar protein aggregation. These results indicate that Bri2 BRICHOS affects qualitatively different aspects of protein misfolding and toxicity via different quaternary structures, suggesting a means to generate molecular chaperone diversity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Catarata/patologia , Ataxia Cerebelar/patologia , Angiopatia Amiloide Cerebral Familiar/patologia , Surdez/patologia , Demência/patologia , Glicoproteínas de Membrana/metabolismo , Agregação Patológica de Proteínas/patologia , Proteínas Adaptadoras de Transdução de Sinal , Amiloide/metabolismo , Neuropatias Amiloides Familiares , Dicroísmo Circular , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/ultraestrutura , Microscopia Eletrônica de Transmissão , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/ultraestrutura , Ligação Proteica , Domínios Proteicos/fisiologia , Dobramento de Proteína , Multimerização Proteica/fisiologia , Proteínas Recombinantes
19.
Mol Cell ; 67(2): 322-333.e6, 2017 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-28689658

RESUMO

The proteasome holoenzyme is activated by its regulatory particle (RP) consisting of two subcomplexes, the lid and the base. A key event in base assembly is the formation of a heterohexameric ring of AAA-ATPases, which is guided by at least four RP assembly chaperones in mammals: PAAF1, p28/gankyrin, p27/PSMD9, and S5b. Using cryogenic electron microscopy, we analyzed the non-AAA structure of the p28-bound human RP at 4.5 Å resolution and determined seven distinct conformations of the Rpn1-p28-AAA subcomplex within the p28-bound RP at subnanometer resolutions. Remarkably, the p28-bound AAA ring does not form a channel in the free RP and spontaneously samples multiple "open" and "closed" topologies at the Rpt2-Rpt6 and Rpt3-Rpt4 interfaces. Our analysis suggests that p28 assists the proteolytic core particle to select a specific conformation of the ATPase ring for RP engagement and is released in a shoehorn-like fashion in the last step of the chaperone-mediated proteasome assembly.


Assuntos
Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Microscopia Crioeletrônica , Células HEK293 , Humanos , Proteínas com Domínio LIM/metabolismo , Proteínas com Domínio LIM/ultraestrutura , Modelos Moleculares , Chaperonas Moleculares/ultraestrutura , Complexo de Endopeptidases do Proteassoma/ultraestrutura , Ligação Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas , Proteínas Proto-Oncogênicas/ultraestrutura , Relação Estrutura-Atividade , Fatores de Transcrição/metabolismo , Fatores de Transcrição/ultraestrutura , Transfecção
20.
Mol Biotechnol ; 59(4-5): 117-127, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28324209

RESUMO

CRM197 is a diphtheria toxin (DT) mutant (G52E) which has been used as a carrier protein for conjugate vaccines. However, it still possesses cytotoxicity toward mammalian cells. The goal of this project was to produce a non-toxic and soluble CRM197EK through introduction of triple amino acid substitutions (K51E/G52E/E148K) in Escherichia coli. The expression of CRM197EKTrxHis was optimized and co-expressed with different molecular chaperones. The soluble CRM197EKTrxHis was produced at a high concentration (97.33 ± 17.47 µg/ml) under the optimal condition (induction with 0.1 mM IPTG at 20 °C for 24 h). Cells containing pG-Tf2, expressing trigger factor and GroEL-GroES, accumulated the highest amount of soluble CRM197EKTrxHis at 111.24 ± 10.40 µg/ml after induction for 24 h at 20 °C. The soluble CRM197EKTrxHis still possesses nuclease activity and completely digest λDNA at 25 and 37 °C with 8- and 4-h incubation, respectively. Molecular modeling of diphtheria toxin, CRM197 and CRM197EK indicated that substitutions of two amino acids (K51E/E148K) may cause poor NAD binding, consistent with the lack of toxicity. Therefore, CRM197EK might be used as a new potential carrier protein. However, further in vivo study is required to confirm its roles as functional carrier protein in conjugate vaccines.


Assuntos
Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/química , Clonagem Molecular/métodos , Escherichia coli/fisiologia , Modelos Químicos , Modelos Moleculares , Proteínas de Bactérias/genética , Sítios de Ligação , Simulação por Computador , Engenharia Metabólica/métodos , Chaperonas Moleculares/biossíntese , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/ultraestrutura , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA