Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38942013

RESUMO

The ability of proteins and RNA to coalesce into phase-separated assemblies, such as the nucleolus and stress granules, is a basic principle in organizing membraneless cellular compartments. While the constituents of biomolecular condensates are generally well documented, the mechanisms underlying their formation under stress are only partially understood. Here, we show in yeast that covalent modification with the ubiquitin-like modifier Urm1 promotes the phase separation of a wide range of proteins. We find that the drop in cellular pH induced by stress triggers Urm1 self-association and its interaction with both target proteins and the Urm1-conjugating enzyme Uba4. Urmylation of stress-sensitive proteins promotes their deposition into stress granules and nuclear condensates. Yeast cells lacking Urm1 exhibit condensate defects that manifest in reduced stress resilience. We propose that Urm1 acts as a reversible molecular "adhesive" to drive protective phase separation of functionally critical proteins under cellular stress.

2.
FEBS Lett ; 597(13): 1679-1680, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37334940

RESUMO

Photosynthesis uses the energy of sunlight to convert water and atmospheric CO2 into sugars, providing food and oxygen for life. The fixation of atmospheric CO2 in this crucial biological process is mediated by the enzyme Rubisco. The inefficiencies of Rubisco have inspired researchers for decades to explore ways to improve its function with the goal of increasing crop yields [1-4], and more recently to combat global warming [5]. In this graphical review we highlight the challenges involved in engineering plant Rubisco, with a focus on the extensive chaperone requirement for its biogenesis. We discuss strategies for engineering the catalytic properties of Rubisco and for sequestering the enzyme in membraneless compartments to increase CO2 fixation.


Assuntos
Dióxido de Carbono , Ribulose-Bifosfato Carboxilase , Ribulose-Bifosfato Carboxilase/genética , Ribulose-Bifosfato Carboxilase/metabolismo , Fotossíntese , Chaperonas Moleculares/metabolismo , Plantas/metabolismo
3.
Methods Mol Biol ; 2563: 269-296, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36227479

RESUMO

Carboxysomes are large, cytosolic bodies present in all cyanobacteria and many proteobacteria that function as the sites of photosynthetic CO2 fixation by the enzyme ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco). The carboxysome lumen is enriched with Rubisco and carbonic anhydrase (CA). The polyhedral proteinaceous shell allows the passage of HCO3- ions into the carboxysome, where they are converted to CO2 by CA. Thus, the carboxysome functions as a CO2-concentrating mechanism (CCM), enhancing the efficiency of Rubisco in CO2 fixation. In ß-cyanobacteria, carboxysome biogenesis first involves the aggregation of Rubisco by CcmM, a scaffolding protein that exists in two isoforms. Both isoforms contain a minimum of three Rubisco small subunit-like (SSUL) domains, connected by flexible linkers. Multivalent interaction between these linked SSUL domains with Rubisco results in phase separation and condensate formation. Here, we use Rubisco and the short isoform of CcmM (M35) of the ß-cyanobacterium Synechococcus elongatus PCC7942 to describe the methods used for in vitro analysis of the mechanism of condensate formation driven by the SSUL domains. The methods include turbidity assays, bright-field and fluorescence microscopy, as well as transmission electron microscopy (TEM) in both negative staining and cryo-conditions.


Assuntos
Anidrases Carbônicas , Ribulose-Bifosfato Carboxilase , Proteínas de Bactérias/metabolismo , Dióxido de Carbono/metabolismo , Anidrases Carbônicas/metabolismo , Organelas/metabolismo , Oxigenases/metabolismo , Isoformas de Proteínas/metabolismo , Ribulose-Bifosfato Carboxilase/metabolismo
4.
Nat Struct Mol Biol ; 28(11): 909-922, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34759380

RESUMO

Carboxysomes in cyanobacteria enclose the enzymes Rubisco and carbonic anhydrase to optimize photosynthetic carbon fixation. Understanding carboxysome assembly has implications in agricultural biotechnology. Here we analyzed the role of the scaffolding protein CcmM of the ß-cyanobacterium Synechococcus elongatus PCC 7942 in sequestrating the hexadecameric Rubisco and the tetrameric carbonic anhydrase, CcaA. We find that the trimeric CcmM, consisting of γCAL oligomerization domains and linked small subunit-like (SSUL) modules, plays a central role in mediation of pre-carboxysome condensate formation through multivalent, cooperative interactions. The γCAL domains interact with the C-terminal tails of the CcaA subunits and additionally mediate a head-to-head association of CcmM trimers. Interestingly, SSUL modules, besides their known function in recruiting Rubisco, also participate in intermolecular interactions with the γCAL domains, providing further valency for network formation. Our findings reveal the mechanism by which CcmM functions as a central organizer of the pre-carboxysome multiprotein matrix, concentrating the core components Rubisco and CcaA before ß-carboxysome shell formation.


Assuntos
Proteínas de Bactérias/metabolismo , Anidrases Carbônicas/metabolismo , Domínios e Motivos de Interação entre Proteínas/fisiologia , Ribulose-Bifosfato Carboxilase/metabolismo , Synechococcus/metabolismo , Condensados Biomoleculares/fisiologia , Microscopia Crioeletrônica , Fotossíntese/fisiologia , Conformação Proteica , Synechococcus/genética
5.
Mol Cell ; 81(14): 2914-2928.e7, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34107307

RESUMO

Molecular chaperones assist with protein folding by interacting with nascent polypeptide chains (NCs) during translation. Whether the ribosome can sense chaperone defects and, in response, abort translation of misfolding NCs has not yet been explored. Here we used quantitative proteomics to investigate the ribosome-associated chaperone network in E. coli and the consequences of its dysfunction. Trigger factor and the DnaK (Hsp70) system are the major NC-binding chaperones. HtpG (Hsp90), GroEL, and ClpB contribute increasingly when DnaK is deficient. Surprisingly, misfolding because of defects in co-translational chaperone function or amino acid analog incorporation results in recruitment of the non-canonical release factor RF3. RF3 recognizes aberrant NCs and then moves to the peptidyltransferase site to cooperate with RF2 in mediating chain termination, facilitating clearance by degradation. This function of RF3 reduces the accumulation of misfolded proteins and is critical for proteostasis maintenance and cell survival under conditions of limited chaperone availability.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Chaperonas Moleculares/metabolismo , Biossíntese de Proteínas/fisiologia , Aminoácidos/metabolismo , Sobrevivência Celular/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Fatores de Terminação de Peptídeos/metabolismo , Peptidil Transferases/metabolismo , Ligação Proteica/fisiologia , Dobramento de Proteína , Proteômica/métodos , Proteostase/fisiologia , Ribossomos/metabolismo
6.
Cell ; 183(2): 457-473.e20, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32979320

RESUMO

Rubisco, the key enzyme of CO2 fixation in photosynthesis, is prone to inactivation by inhibitory sugar phosphates. Inhibited Rubisco undergoes conformational repair by the hexameric AAA+ chaperone Rubisco activase (Rca) in a process that is not well understood. Here, we performed a structural and mechanistic analysis of cyanobacterial Rca, a close homolog of plant Rca. In the Rca:Rubisco complex, Rca is positioned over the Rubisco catalytic site under repair and pulls the N-terminal tail of the large Rubisco subunit (RbcL) into the hexamer pore. Simultaneous displacement of the C terminus of the adjacent RbcL opens the catalytic site for inhibitor release. An alternative interaction of Rca with Rubisco is mediated by C-terminal domains that resemble the small Rubisco subunit. These domains, together with the N-terminal AAA+ hexamer, ensure that Rca is packaged with Rubisco into carboxysomes. The cyanobacterial Rca is a dual-purpose protein with functions in Rubisco repair and carboxysome organization.


Assuntos
Cianobactérias/metabolismo , Ribulose-Bifosfato Carboxilase/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Organelas/metabolismo , Fotossíntese/fisiologia , Ribulose-Bifosfato Carboxilase/fisiologia , Ativador de Plasminogênio Tecidual/química , Ativador de Plasminogênio Tecidual/metabolismo
7.
FEBS Lett ; 594(17): 2770-2781, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32446288

RESUMO

Molecular chaperones are highly conserved proteins that promote proper folding of other proteins in vivo. Diverse chaperone systems assist de novo protein folding and trafficking, the assembly of oligomeric complexes, and recovery from stress-induced unfolding. A fundamental function of molecular chaperones is to inhibit unproductive protein interactions by recognizing and protecting hydrophobic surfaces that are exposed during folding or following proteotoxic stress. Beyond this basic principle, it is now clear that chaperones can also actively and specifically accelerate folding reactions in an ATP-dependent manner. We focus on the bacterial Hsp70 and chaperonin systems as paradigms, and review recent work that has advanced our understanding of how these chaperones act as catalysts of protein folding.


Assuntos
Trifosfato de Adenosina/química , Chaperonina 60/química , Chaperoninas/química , Proteínas de Escherichia coli/química , Escherichia coli/genética , Proteínas de Choque Térmico HSP70/química , Chaperonas Moleculares/química , Trifosfato de Adenosina/metabolismo , Biocatálise , Chaperonina 60/genética , Chaperonina 60/metabolismo , Chaperoninas/genética , Chaperoninas/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Desdobramento de Proteína
8.
Trends Biochem Sci ; 45(9): 748-763, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32471779

RESUMO

A major challenge faced by human civilization is to ensure that agricultural productivity keeps pace with population growth and a changing climate. All food supply is generated, directly or indirectly, through the process of photosynthesis, with the enzyme ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) catalyzing the assimilation of atmospheric CO2. Despite its pivotal role, Rubisco is a remarkably inefficient enzyme and must be made by plants in large quantities. However, efforts to enhance Rubisco performance by bioengineering have been hampered by its extensive reliance on molecular chaperones and auxiliary factors for biogenesis, metabolic repair, and packaging into membraneless microcompartments. Here, we review recent advances in understanding these complex machineries and discuss their implications for improving Rubisco carboxylase activity with the goal to increase crop yields.


Assuntos
Chaperonas Moleculares , Plantas/enzimologia , Ribulose-Bifosfato Carboxilase , Chaperonas Moleculares/metabolismo , Fotossíntese , Ribulose-Bifosfato Carboxilase/metabolismo
9.
PLoS One ; 15(4): e0230090, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32339190

RESUMO

Chaperonins are ubiquitous molecular chaperones found in all domains of life. They form ring-shaped complexes that assist in the folding of substrate proteins in an ATP-dependent reaction cycle. Key to the folding cycle is the transient encapsulation of substrate proteins by the chaperonin. Here we present a structural and functional characterization of the chaperonin gp146 (ɸEL) from the phage EL of Pseudomonas aeruginosa. ɸEL, an evolutionarily distant homolog of bacterial GroEL, is active in ATP hydrolysis and prevents the aggregation of denatured protein in a nucleotide-dependent manner. However, ɸEL failed to refold the encapsulation-dependent model substrate rhodanese and did not interact with E. coli GroES, the lid-shaped co-chaperone of GroEL. ɸEL forms tetradecameric double-ring complexes, which dissociate into single rings in the presence of ATP. Crystal structures of ɸEL (at 3.54 and 4.03 Å) in presence of ATP•BeFx revealed two distinct single-ring conformational states, both with open access to the ring cavity. One state showed uniform ATP-bound subunit conformations (symmetric state), whereas the second combined distinct ATP- and ADP-bound subunit conformations (asymmetric state). Cryo-electron microscopy of apo-ɸEL revealed a double-ring structure composed of rings in the asymmetric state (3.45 Å resolution). We propose that the phage chaperonin undergoes nucleotide-dependent conformational switching between double- and single rings and functions in aggregation prevention without substrate protein encapsulation. Thus, ɸEL may represent an evolutionarily more ancient chaperonin prior to acquisition of the encapsulation mechanism.


Assuntos
Chaperoninas/química , Dobramento de Proteína , Fagos de Pseudomonas/química , Pseudomonas aeruginosa/virologia , Proteínas Virais/química , Chaperonina 10/química , Chaperonina 60/química , Microscopia Crioeletrônica , Escherichia coli/química , Proteínas de Escherichia coli/química , Domínios Proteicos , Fagos de Pseudomonas/metabolismo
10.
J Mol Biol ; 432(7): 2304-2318, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32135190

RESUMO

The cylindrical chaperonin GroEL and its cofactor GroES mediate ATP-dependent protein folding in Escherichia coli by transiently encapsulating non-native substrate in a nano-cage formed by the GroEL ring cavity and the lid-shaped GroES. Mechanistic studies of GroEL/ES with heterologous protein substrates suggested that the chaperonin is inefficient, typically requiring multiple ATP-dependent encapsulation cycles with only a few percent of protein folded per cycle. Here we analyzed the spontaneous and chaperonin-assisted folding of the essential enzyme 5,10-methylenetetrahydrofolate reductase (MetF) of E. coli, an obligate GroEL/ES substrate. We found that MetF, a homotetramer of 33-kDa subunits with (ß/α)8 TIM-barrel fold, populates a kinetically trapped folding intermediate(s) (MetF-I) upon dilution from denaturant that fails to convert to the native state, even in the absence of aggregation. GroEL/ES recognizes MetF-I and catalyzes rapid folding, with ~50% of protein folded in a single round of encapsulation. Analysis by hydrogen/deuterium exchange at peptide resolution showed that the MetF subunit folds to completion in the GroEL/ES nano-cage and binds its cofactor flavin adenine dinucleotide. Rapid folding required the net negative charge character of the wall of the chaperonin cavity. These findings reveal a remarkable capacity of GroEL/ES to catalyze folding of an endogenous substrate protein that would have coevolved with the chaperonin system.


Assuntos
5,10-Metilenotetra-Hidrofolato Redutase (FADH2)/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Dobramento de Proteína , Trifosfato de Adenosina/metabolismo , Catálise , Cinética , Modelos Moleculares , Conformação Proteica , Termodinâmica
11.
Nat Commun ; 11(1): 365, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31953415

RESUMO

The ATP-dependent Hsp70 chaperones (DnaK in E. coli) mediate protein folding in cooperation with J proteins and nucleotide exchange factors (E. coli DnaJ and GrpE, respectively). The Hsp70 system prevents protein aggregation and increases folding yields. Whether it also enhances the rate of folding remains unclear. Here we show that DnaK/DnaJ/GrpE accelerate the folding of the multi-domain protein firefly luciferase (FLuc) ~20-fold over the rate of spontaneous folding measured in the absence of aggregation. Analysis by single-pair FRET and hydrogen/deuterium exchange identified inter-domain misfolding as the cause of slow folding. DnaK binding expands the misfolded region and thereby resolves the kinetically-trapped intermediates, with folding occurring upon GrpE-mediated release. In each round of release DnaK commits a fraction of FLuc to fast folding, circumventing misfolding. We suggest that by resolving misfolding and accelerating productive folding, the bacterial Hsp70 system can maintain proteins in their native states under otherwise denaturing stress conditions.


Assuntos
Proteínas de Escherichia coli/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Domínios Proteicos , Dobramento de Proteína , Animais , Sítios de Ligação , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Vaga-Lumes/enzimologia , Vaga-Lumes/genética , Proteínas de Choque Térmico HSP40/química , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico/química , Cinética , Luciferases de Vaga-Lume/química , Luciferases de Vaga-Lume/genética , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Conformação Proteica
12.
Cell Rep ; 28(5): 1335-1345.e6, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31365874

RESUMO

Stress-inducible molecular chaperones have essential roles in maintaining protein homeostasis, but the extent to which they affect overall proteome stability remains unclear. Here, we analyze the effects of the DnaK (Hsp70) system on protein stability in Escherichia coli using pulse proteolysis combined with quantitative proteomics. We quantify ∼1,500 soluble proteins and find ∼500 of these to be protease sensitive under normal growth conditions, indicating a high prevalence of conformationally dynamic proteins, forming a metastable subproteome. Acute heat stress results in the unfolding of an additional ∼200 proteins, reflected in the exposure of otherwise buried hydrophobic regions. Overexpression of the DnaK chaperone system markedly stabilizes numerous thermo-sensitive proteins, including multiple ribosomal proteins and large, hetero-oligomeric proteins containing the evolutionarily ancient c.37 fold (P loop nucleoside triphosphate hydrolases). Thus, the Hsp70 system, in addition to its known chaperone functions, has a remarkable capacity to stabilize proteins in their folded states under denaturing stress conditions.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Resposta ao Choque Térmico/fisiologia , Proteoma/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Choque Térmico HSP70/genética , Proteoma/genética , Proteômica
13.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 4): 278-289, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30950829

RESUMO

Phosphoribulokinase (PRK) catalyses the ATP-dependent phosphorylation of ribulose 5-phosphate to give ribulose 1,5-bisphosphate. Regulation of this reaction in response to light controls carbon fixation during photosynthesis. Here, the crystal structure of PRK from the cyanobacterium Synechococcus sp. strain PCC 6301 is presented. The enzyme is dimeric and has an α/ß-fold with an 18-stranded ß-sheet at its core. Interestingly, a disulfide bond is found between Cys40 and the P-loop residue Cys18, revealing the structural basis for the redox inactivation of PRK activity. A second disulfide bond appears to rigidify the dimer interface and may thereby contribute to regulation by the adaptor protein CP12 and glyceraldehyde-3-phosphate dehydrogenase.


Assuntos
Fosfotransferases (Aceptor do Grupo Álcool)/química , Synechococcus/enzimologia , Sequência de Aminoácidos , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Funções Verossimilhança , Modelos Moleculares , Filogenia , Multimerização Proteica , Homologia Estrutural de Proteína
14.
FEBS Lett ; 593(6): 611-621, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30815863

RESUMO

Improving the performance of the key photosynthetic enzyme Ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) by protein engineering is a critical strategy for increasing crop yields. The extensive chaperone requirement of plant Rubisco for folding and assembly has long been an impediment to this goal. Production of plant Rubisco in Escherichia coli requires the coexpression of the chloroplast chaperonin and four assembly factors. Here, we demonstrate that simultaneous expression of Rubisco and chaperones from a T7 promotor produces high levels of functional enzyme. Expressing the small subunit of Rubisco with a C-terminal hexahistidine-tag further improved assembly, resulting in a ~ 12-fold higher yield than the previously published procedure. The expression system described here provides a platform for the efficient production and engineering of plant Rubisco.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/enzimologia , Clonagem Molecular/métodos , Chaperoninas do Grupo I/genética , Chaperonas Moleculares/genética , Proteínas de Ligação a Fosfato/genética , Ribulose-Bifosfato Carboxilase/genética , Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Dióxido de Carbono/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Chaperoninas do Grupo I/metabolismo , Histidina/genética , Histidina/metabolismo , Cinética , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Fotossíntese/genética , Dobramento de Proteína , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Subunidades Proteicas/genética , Subunidades Proteicas/isolamento & purificação , Subunidades Proteicas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Ribulose-Bifosfato Carboxilase/isolamento & purificação , Ribulose-Bifosfato Carboxilase/metabolismo
16.
Nature ; 563(7730): 209-213, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30232455

RESUMO

Tc toxins secrete toxic enzymes into host cells using a unique syringe-like injection mechanism. They are composed of three subunits, TcA, TcB and TcC. TcA forms the translocation channel and the TcB-TcC heterodimer functions as a cocoon that shields the toxic enzyme. Binding of the cocoon to the channel triggers opening of the cocoon and translocation of the toxic enzyme into the channel. Here we show in atomic detail how the assembly of the three components activates the toxin. We find that part of the cocoon completely unfolds and refolds into an alternative conformation upon binding. The presence of the toxic enzyme inside the cocoon is essential for its subnanomolar binding affinity for the TcA subunit. The enzyme passes through a narrow negatively charged constriction site inside the cocoon, probably acting as an extruder that releases the unfolded protein with its C terminus first into the translocation channel.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Microscopia Crioeletrônica , Complexos Multiproteicos/ultraestrutura , Photorhabdus/ultraestrutura , Redobramento de Proteína , Desdobramento de Proteína , ADP Ribose Transferases/química , ADP Ribose Transferases/metabolismo , ADP Ribose Transferases/ultraestrutura , Toxinas Bacterianas/biossíntese , Citotoxinas/biossíntese , Citotoxinas/química , Citotoxinas/metabolismo , Modelos Biológicos , Modelos Moleculares , Complexos Multiproteicos/biossíntese , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Photorhabdus/química , Conformação Proteica , Transporte Proteico
17.
Cell ; 174(6): 1507-1521.e16, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30100183

RESUMO

The hetero-oligomeric chaperonin of eukarya, TRiC, is required to fold the cytoskeletal protein actin. The simpler bacterial chaperonin system, GroEL/GroES, is unable to mediate actin folding. Here, we use spectroscopic and structural techniques to determine how TRiC promotes the conformational progression of actin to the native state. We find that actin fails to fold spontaneously even in the absence of aggregation but populates a kinetically trapped, conformationally dynamic state. Binding of this frustrated intermediate to TRiC specifies an extended topology of actin with native-like secondary structure. In contrast, GroEL stabilizes bound actin in an unfolded state. ATP binding to TRiC effects an asymmetric conformational change in the chaperonin ring. This step induces the partial release of actin, priming it for folding upon complete release into the chaperonin cavity, mediated by ATP hydrolysis. Our results reveal how the unique features of TRiC direct the folding pathway of an obligate eukaryotic substrate.


Assuntos
Actinas/metabolismo , Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Actinas/química , Trifosfato de Adenosina/metabolismo , Animais , Bovinos , Chaperonina 10/química , Chaperonina 60/química , Microscopia Crioeletrônica , Desoxirribonuclease I/química , Desoxirribonuclease I/metabolismo , Medição da Troca de Deutério , Humanos , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína
18.
Biochemistry ; 57(23): 3210-3216, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29589905

RESUMO

Ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco), a ∼530 kDa complex of 8 large (RbcL) and 8 small subunits (RbcS), mediates the fixation of atmospheric CO2 into usable sugars during photosynthesis. Despite its fundamental role, Rubisco is a remarkably inefficient enzyme and thus is produced by plants in huge amounts. It has long been a key target for bioengineering with the goal to increase crop yields. However, such efforts have been hampered by the complex requirement of Rubisco biogenesis for molecular chaperones. Recent studies have identified an array of auxiliary factors needed for the folding and assembly of the Rubisco subunits. The folding of plant RbcL subunits is mediated by the cylindrical chloroplast chaperonin, Cpn60, and its cofactor Cpn20. Folded RbcL requires a number of additional Rubisco specific assembly chaperones, including RbcX, Rubisco accumulation factors 1 (Raf1) and 2 (Raf2), and the Bundle sheath defective-2 (BSD2), to mediate the assembly of the RbcL8 intermediate complex. Incorporation of the RbcS and displacement of the assembly factors generates the active holoenzyme. An Escherichia coli strain expressing the chloroplast chaperonin and auxiliary factors now allows the expression of functional plant Rubisco, paving the way for Rubisco engineering by large scale mutagenesis. Here, we review our current understanding on how these chaperones cooperate to produce one of the most important enzymes in nature.


Assuntos
Escherichia coli , Chaperonas Moleculares , Ribulose-Bifosfato Carboxilase , Escherichia coli/genética , Escherichia coli/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Ribulose-Bifosfato Carboxilase/biossíntese , Ribulose-Bifosfato Carboxilase/genética
19.
Cell ; 172(3): 605-617.e11, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29336887

RESUMO

The bacterial chaperonin GroEL and its cofactor, GroES, form a nano-cage for a single molecule of substrate protein (SP) to fold in isolation. GroEL and GroES undergo an ATP-regulated interaction cycle to close and open the folding cage. GroEL consists of two heptameric rings stacked back to back. Here, we show that GroEL undergoes transient ring separation, resulting in ring exchange between complexes. Ring separation occurs upon ATP-binding to the trans ring of the asymmetric GroEL:7ADP:GroES complex in the presence or absence of SP and is a consequence of inter-ring negative allostery. We find that a GroEL mutant unable to perform ring separation is folding active but populates symmetric GroEL:GroES2 complexes, where both GroEL rings function simultaneously rather than sequentially. As a consequence, SP binding and release from the folding chamber is inefficient, and E. coli growth is impaired. We suggest that transient ring separation is an integral part of the chaperonin mechanism.


Assuntos
Chaperonina 60/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Chaperonina 10/metabolismo , Chaperonina 60/química , Chaperonina 60/genética , Mutação , Ligação Proteica
20.
Protein Sci ; 26(12): 2324-2333, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28960553

RESUMO

Ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) mediates the fixation of atmospheric CO2 in photosynthesis by catalyzing the carboxylation of the 5-carbon sugar ribulose-1,5-bisphosphate (RuBP). Despite its pivotal role, Rubisco is an inefficient enzyme and thus has been a key target for bioengineering. However, efforts to increase crop yields by Rubisco engineering remain unsuccessful, due in part to the complex machinery of molecular chaperones required for Rubisco biogenesis and metabolic repair. While the large subunit of Rubisco generally requires the chaperonin system for folding, the evolution of the hexadecameric Rubisco from its dimeric precursor resulted in the dependence on an array of additional factors required for assembly. Moreover, Rubisco function can be inhibited by a range of sugar-phosphate ligands. Metabolic repair of Rubisco depends on remodeling by the ATP-dependent Rubisco activase and hydrolysis of inhibitors by specific phosphatases. This review highlights our work toward understanding the structure and mechanism of these auxiliary machineries.


Assuntos
Chaperoninas , Ribulose-Bifosfato Carboxilase , Proteínas de Bactérias , Modelos Moleculares , Dobramento de Proteína , Multimerização Proteica , Rhodobacter
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...