Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 512
1.
Protein Expr Purif ; 192: 106041, 2022 04.
Article En | MEDLINE | ID: mdl-34953978

The gene encoding the cAMP-dependent protein kinase (PKA) catalytic subunit-like protein PKAC1 from the Venezuelan TeAp-N/D1 strain of Trypanosoma equiperdum was cloned, and the recombinant TeqPKAC1 protein was overexpressed in bacteria. A major polypeptide with an apparent molecular mass of ∼38 kDa was detected by SDS-polyacrylamide gel electrophoresis, and immunoblotting using antibodies against the human PKA catalytic subunit α. Unfortunately, most of the expressed TeqPKAC1 was highly insoluble. Polypeptides of 36-38 kDa and 45-50 kDa were predominantly seen by immunoblotting in the bacterial particulate and cytosolic fractions, respectively. Since the incorporation of either 4% Triton X-100 or 3% sarkosyl or a mixture of 10 mM MgCl2 and 1 mM ATP (MgATP) improved the solubilization of TeqPKAC1, we used a combination of Triton X-100, sarkosyl and MgATP to solubilize the recombinant protein. TeqPKAC1 was purified by first reconstituting a hybrid holoenzyme between the recombinant protein and a mammalian poly-His-tagged PKA regulatory subunit that was immobilized on a Ni2+-chelating affinity resin, and then by eluting TeqPKAC1 using cAMP. TeqPKAC1 was functional given that it was capable of phosphorylating PKA catalytic subunit substrates, such as kemptide (LRRASLG), histone type II-AS, and the peptide SP20 (TTYADFIASGRTGRRNSIHD), and was inhibited by the peptide IP20 (TTYADFIASGRTGRRNAIHD), which contains the inhibitory motif of the PKA-specific heat-stable inhibitor PKI-α. Optimal enzymatic activity was obtained at 37 °C and pH 8.0-9.0; and the order of effectiveness of nucleotide triphosphates and divalent cations was ATP ¼ GTP â‰… ITP and Mg2+ â‰… Mn2+ â‰… Fe2+ ¼ Ca2+ â‰… Zn2, respectively.


Cloning, Molecular , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Trypanosoma/enzymology , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/isolation & purification , Phosphorylation , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility , Trypanosoma/chemistry , Trypanosoma/genetics
2.
Molecules ; 26(21)2021 Oct 27.
Article En | MEDLINE | ID: mdl-34770894

Phycoerythrin is a major light-harvesting pigment of red algae and cyanobacteria that is widely used as a fluorescent probe or as a colorant in the food and cosmetic industries. In this study, phycoerythrin was extracted from the red algae Pyropia yezoensis and purified by ammonium sulfate precipitation and various chromatography methods. The purified phycoerythrin was analyzed by UV-visible and fluorescence spectroscopy. The isolated pigment had the typical spectrum of R-phycoerythrin, with a trimmer state with absorbance maxima at 497, 536, and 565 nm. It was further purified and identified by LC-MS/MS and Mascot search. It showed a 100% sequence similarity with the R-phycoerythrin alpha subunit of Pyropia yezoensis. The molecular mass was 17.97 kDa. The antioxidant activity of the purified R-phycoerythrin alpha subunit was analyzed. It showed significant antioxidant activity in ABTS and FRAP assays and had significant cytotoxicity against HepG2 cells.


Aquatic Organisms/chemistry , Biological Products/chemistry , Biological Products/pharmacology , Phycoerythrin/chemistry , Protein Subunits/chemistry , Protein Subunits/pharmacology , Rhodophyta/chemistry , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/isolation & purification , Antioxidants/pharmacology , Biological Products/isolation & purification , Cell Line, Tumor , Cell Survival/drug effects , Chemical Fractionation/methods , Chromatography, Liquid , Dose-Response Relationship, Drug , Humans , Peptide Fragments , Protein Subunits/isolation & purification , Tandem Mass Spectrometry
3.
Proteins ; 89(5): 477-482, 2021 05.
Article En | MEDLINE | ID: mdl-33378096

The mitochondrial F1 FO -ATPase in the presence of the natural cofactor Mg2+ acts as the enzyme of life by synthesizing ATP, but it can also hydrolyze ATP to pump H+ . Interestingly, Mg2+ can be replaced by Ca2+ , but only to sustain ATP hydrolysis and not ATP synthesis. When Ca2+ inserts in F1 , the torque generation built by the chemomechanical coupling between F1 and the rotating central stalk was reported as unable to drive the transmembrane H+ flux within FO . However, the failed H+ translocation is not consistent with the oligomycin-sensitivity of the Ca2+ -dependent F1 FO -ATP(hydrol)ase. New enzyme roles in mitochondrial energy transduction are suggested by recent advances. Accordingly, the structural F1 FO -ATPase distortion driven by ATP hydrolysis sustained by Ca2+ is consistent with the permeability transition pore signal propagation pathway. The Ca2+ -activated F1 FO -ATPase, by forming the pore, may contribute to dissipate the transmembrane H+ gradient created by the same enzyme complex.


Adenosine Triphosphate/chemistry , Calcium/chemistry , Coenzymes/chemistry , Magnesium/chemistry , Mitochondria, Heart/chemistry , Mitochondrial Proton-Translocating ATPases/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Calcium/metabolism , Cations, Divalent , Coenzymes/metabolism , Hydrolysis/drug effects , Kinetics , Magnesium/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Permeability Transition Pore/chemistry , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/isolation & purification , Mitochondrial Proton-Translocating ATPases/metabolism , Models, Molecular , Myocardium/chemistry , Myocardium/enzymology , Oligomycins/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Substrate Specificity , Swine , Thermodynamics
4.
Methods Mol Biol ; 2247: 3-16, 2021.
Article En | MEDLINE | ID: mdl-33301109

Membrane proteins constitute an important class of proteins for medical, pharmaceutical, and biotechnological reasons. Understanding the structure and function of membrane proteins and their complexes is of key importance, but the progress in this area is slow because of the difficulties to produce them in sufficient quality and quantity. Overexpression of membrane proteins is often restricted by the limited capability of translocation systems to integrate proteins into the membrane and to fold them properly. Purification of membrane proteins requires their isolation from the membrane, which is a further challenge. The choice of expression system, detergents, and purification tags is therefore an important decision. Here, we present a protocol for expression in bacteria and isolation of a seven-subunit membrane protein complex, the bacterial holo-translocon, which can serve as a starting point for the production of other membrane protein complexes for structural and functional studies.


Membrane Proteins/biosynthesis , Membrane Proteins/isolation & purification , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/isolation & purification , Protein Subunits/biosynthesis , Protein Subunits/isolation & purification , Recombinant Proteins , Chromatography, Affinity , Chromatography, Gel , Escherichia coli/genetics , Gene Expression , Membrane Proteins/chemistry , Plasmids , Promoter Regions, Genetic , Protein Multimerization , Protein Subunits/chemistry
5.
J Vis Exp ; (163)2020 09 26.
Article En | MEDLINE | ID: mdl-33044464

Structural studies with tryptophan synthase (TS) bienzyme complex (α2ß2 TS) from Salmonella typhimurium have been performed to better understand its catalytic mechanism, allosteric behavior, and details of the enzymatic transformation of substrate to product in PLP-dependent enzymes. In this work, a novel expression system to produce the isolated α- and isolated ß-subunit allowed the purification of high amounts of pure subunits and α2ß2 StTS complex from the isolated subunits within 2 days. Purification was carried out by affinity chromatography followed by cleavage of the affinity tag, ammonium sulfate precipitation, and size exclusion chromatography (SEC). To better understand the role of key residues at the enzyme ß-site, site-direct mutagenesis was performed in prior structural studies. Another protocol was created to purify the wild type and mutant α2ß2 StTS complexes. A simple, fast and efficient protocol using ammonium sulfate fractionation and SEC allowed purification of α2ß2 StTS complex in a single day. Both purification protocols described in this work have considerable advantages when compared with previous protocols to purify the same complex using PEG 8000 and spermine to crystalize the α2ß2 StTS complex along the purification protocol. Crystallization of wild type and some mutant forms occurs under slightly different conditions, impairing the purification of some mutants using PEG 8000 and spermine. To prepare crystals suitable for x-ray crystallographic studies several efforts were made to optimize crystallization, crystal quality and cryoprotection. The methods presented here should be generally applicable for purification of tryptophan synthase subunits and wild type and mutant α2ß2 StTS complexes.


Mutagenesis, Site-Directed/methods , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Polymerase Chain Reaction/methods , Tryptophan Synthase/genetics , Tryptophan Synthase/isolation & purification , Catalysis , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/metabolism , Protein Subunits/isolation & purification , Recombinant Proteins/metabolism , Reproducibility of Results , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Static Electricity , Tryptophan Synthase/chemistry
6.
Commun Biol ; 3(1): 431, 2020 08 07.
Article En | MEDLINE | ID: mdl-32770029

rnf genes are widespread in bacteria and biochemical and genetic data are in line with the hypothesis that they encode a membrane-bound enzyme that oxidizes reduced ferredoxin and reduces NAD and vice versa, coupled to ion transport across the cytoplasmic membrane. The Rnf complex is of critical importance in many bacteria for energy conservation but also for reverse electron transport to drive ferredoxin reduction. However, the enzyme has never been purified and thus, ion transport could not be demonstrated yet. Here, we have purified the Rnf complex from the anaerobic, fermenting thermophilic bacterium Thermotoga maritima and show that is a primary Na+ pump. These studies provide the proof that the Rnf complex is indeed an ion (Na+) translocating, respiratory enzyme. Together with a Na+-F1FO ATP synthase it builds a simple, two-limb respiratory chain in T. maritima. The physiological role of electron transport phosphorylation in a fermenting bacterium is discussed.


Bacterial Proteins/metabolism , Fermentation , Sodium/metabolism , Thermotoga maritima/enzymology , Adenosine Triphosphate/metabolism , Bacterial Proteins/isolation & purification , Dicyclohexylcarbodiimide/pharmacology , Fermentation/drug effects , Ferredoxins/metabolism , Glucose/metabolism , Hydrolysis , Ion Transport/drug effects , Ionophores/pharmacology , Liposomes , Models, Biological , Oxidoreductases/metabolism , Protein Subunits/isolation & purification , Proton-Translocating ATPases/isolation & purification , Proton-Translocating ATPases/metabolism , Thermotoga maritima/drug effects
7.
Endocrinology ; 161(8)2020 08 01.
Article En | MEDLINE | ID: mdl-32569368

Ovarian-derived inhibin A and inhibin B (heterodimers of common α- and differing ß-subunits) are secreted throughout the menstrual cycle in a discordant pattern, with smaller follicles producing inhibin B, whereas the dominant follicle and corpus luteum produce inhibin A. The classical function for endocrine inhibins is to block signalling by activins (homodimers of ß-subunits) in gonadotrope cells of the anterior pituitary and, thereby, inhibit the synthesis of FSH. Whether inhibin A and inhibin B have additional physiological functions is unknown, primarily because producing sufficient quantities of purified inhibins, in the absence of contaminating activins, for preclinical studies has proven extremely difficult. Here, we describe novel methodology to enhance inhibin A and inhibin B activity and to produce these ligands free of contaminating activins. Using computational modeling and targeted mutagenesis, we identified a point mutation in the activin ß A-subunit, A347H, which completely disrupted activin dimerization and activity. Importantly, this ß A-subunit mutation had minimal effect on inhibin A bioactivity. Mutation of the corresponding residue in the inhibin ß B-subunit, G329E, similarly disrupted activin B synthesis/activity without affecting inhibin B production. Subsequently, we enhanced inhibin A potency by modifying the binding site for its co-receptor, betaglycan. Introducing a point mutation into the α-subunit (S344I) increased inhibin A potency ~12-fold. This study has identified a means to eliminate activin A/B interference during inhibin A/B production, and has facilitated the generation of potent inhibin A and inhibin B agonists for physiological exploration.


Inhibins , Protein Engineering/methods , Female , HEK293 Cells , Humans , Inhibins/genetics , Inhibins/isolation & purification , Inhibins/metabolism , Inhibins/pharmacology , Membrane Proteins , Models, Molecular , Mutagenesis/physiology , Ovary/metabolism , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Protein Multimerization/genetics , Protein Structure, Quaternary/genetics , Protein Structure, Tertiary/genetics , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Protein Subunits/pharmacology , Saccharomyces cerevisiae Proteins , Transfection
8.
Cell ; 180(6): 1130-1143.e20, 2020 03 19.
Article En | MEDLINE | ID: mdl-32160528

Fatty acid synthases (FASs) are central to metabolism but are also of biotechnological interest for the production of fine chemicals and biofuels from renewable resources. During fatty acid synthesis, the growing fatty acid chain is thought to be shuttled by the dynamic acyl carrier protein domain to several enzyme active sites. Here, we report the discovery of a γ subunit of the 2.6 megadalton α6-ß6S. cerevisiae FAS, which is shown by high-resolution structures to stabilize a rotated FAS conformation and rearrange ACP domains from equatorial to axial positions. The γ subunit spans the length of the FAS inner cavity, impeding reductase activities of FAS, regulating NADPH turnover by kinetic hysteresis at the ketoreductase, and suppressing off-pathway reactions at the enoylreductase. The γ subunit delineates the functional compartment within FAS. As a scaffold, it may be exploited to incorporate natural and designed enzymatic activities that are not present in natural FAS.


Fatty Acid Synthases/chemistry , Fatty Acid Synthases/metabolism , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Acyltransferases/metabolism , Binding Sites , Catalytic Domain , Cryoelectron Microscopy/methods , Crystallography, X-Ray/methods , Fatty Acids/biosynthesis , Fatty Acids/chemistry , Models, Molecular , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
9.
Methods Mol Biol ; 2127: 1-11, 2020.
Article En | MEDLINE | ID: mdl-32112311

Saccharomyces cerevisiae is a useful eukaryotic expression system for mitochondrial membrane proteins due to its ease of growth and ability to provide a native membrane environment. The development of the pBEVY vector system has further increased the potential of S. cerevisiae as an expression system by creating a method for expressing multiple proteins simultaneously. This vector system is amenable to the expression and purification of multi-subunit protein complexes. Here we describe the cloning, yeast transformation, and co-expression of multi-subunit outer mitochondrial membrane complexes using the pBEVY vector system.


Cloning, Molecular/methods , Membrane Proteins , Mitochondrial Membranes/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Fractionation/methods , Gene Expression Regulation, Fungal , Genetic Vectors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Membranes/chemistry , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/isolation & purification , Mitochondrial Proteins/metabolism , Organisms, Genetically Modified , Protein Multimerization/genetics , Protein Processing, Post-Translational , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transformation, Genetic
10.
Nat Commun ; 10(1): 5823, 2019 12 20.
Article En | MEDLINE | ID: mdl-31862883

Purified mitochondrial ATP synthase has been shown to form Ca2+-activated, large conductance channel activity similar to that of mitochondrial megachannel (MMC) or mitochondrial permeability transition pore (mPTP) but the oligomeric state required for channel formation is being debated. We reconstitute purified monomeric ATP synthase from porcine heart mitochondria into small unilamellar vesicles (SUVs) with the lipid composition of mitochondrial inner membrane and analyze its oligomeric state by electron cryomicroscopy. The cryo-EM density map reveals the presence of a single ATP synthase monomer with no density seen for a second molecule tilted at an 86o angle relative to the first. We show that this preparation of SUV-reconstituted ATP synthase monomers, when fused into giant unilamellar vesicles (GUVs), forms voltage-gated and Ca2+-activated channels with the key features of mPTP. Based on our findings we conclude that the ATP synthase monomer is sufficient, and dimer formation is not required, for mPTP activity.


Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/ultrastructure , Protein Subunits/metabolism , Animals , Calcium/metabolism , Cryoelectron Microscopy , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proton-Translocating ATPases/isolation & purification , Protein Subunits/isolation & purification , Swine , Unilamellar Liposomes/isolation & purification , Unilamellar Liposomes/metabolism
11.
Sci Rep ; 9(1): 16850, 2019 11 14.
Article En | MEDLINE | ID: mdl-31727948

Recombinant protein expression for structural and therapeutic applications requires the use of systems with high expression yields. Escherichia coli is considered the workhorse for this purpose, given its fast growth rate and feasible manipulation. However, bacterial inclusion body formation remains a challenge for further protein purification. We analyzed and optimized the expression conditions for three different proteins: an anti-MICA scFv, MICA, and p19 subunit of IL-23. We used a response surface methodology based on a three-level Box-Behnken design, which included three factors: post-induction temperature, post-induction time and IPTG concentration. Comparing this information with soluble protein data in a principal component analysis revealed that insoluble and soluble proteins have different optimal conditions for post-induction temperature, post-induction time, IPTG concentration and in amino acid sequence features. Finally, we optimized the refolding conditions of the least expressed protein, anti-MICA scFv, using a fast dilution protocol with different additives, obtaining soluble and active scFv for binding assays. These results allowed us to obtain higher yields of proteins expressed in inclusion bodies. Further studies using the system proposed in this study may lead to the identification of optimal environmental factors for a given protein sequence, favoring the acceleration of bioprocess development and structural studies.


Cloning, Molecular/methods , Escherichia coli/genetics , Histocompatibility Antigens Class I/genetics , Interleukin-23/genetics , Single-Chain Antibodies/genetics , Amino Acid Sequence , Escherichia coli/drug effects , Escherichia coli/metabolism , Factor Analysis, Statistical , Gene Expression/drug effects , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/isolation & purification , Humans , Inclusion Bodies/chemistry , Interleukin-23/chemistry , Interleukin-23/isolation & purification , Isopropyl Thiogalactoside/pharmacology , Principal Component Analysis , Protein Refolding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/isolation & purification , Solubility
12.
Nature ; 576(7786): 321-325, 2019 12.
Article En | MEDLINE | ID: mdl-31597161

Host infection by pathogenic mycobacteria, such as Mycobacterium tuberculosis, is facilitated by virulence factors that are secreted by type VII secretion systems1. A molecular understanding of the type VII secretion mechanism has been hampered owing to a lack of three-dimensional structures of the fully assembled secretion apparatus. Here we report the cryo-electron microscopy structure of a membrane-embedded core complex of the ESX-3/type VII secretion system from Mycobacterium smegmatis. The core of the ESX-3 secretion machine consists of four protein components-EccB3, EccC3, EccD3 and EccE3, in a 1:1:2:1 stoichiometry-which form two identical protomers. The EccC3 coupling protein comprises a flexible array of four ATPase domains, which are linked to the membrane through a stalk domain. The domain of unknown function (DUF) adjacent to the stalk is identified as an ATPase domain that is essential for secretion. EccB3 is predominantly periplasmatic, but a small segment crosses the membrane and contacts the stalk domain. This suggests that conformational changes in the stalk domain-triggered by substrate binding at the distal end of EccC3 and subsequent ATP hydrolysis in the DUF-could be coupled to substrate secretion to the periplasm. Our results reveal that the architecture of type VII secretion systems differs markedly from that of other known secretion machines2, and provide a structural understanding of these systems that will be useful for the design of antimicrobial strategies that target bacterial virulence.


Cryoelectron Microscopy , Mycobacterium smegmatis/chemistry , Type VII Secretion Systems/chemistry , Type VII Secretion Systems/ultrastructure , Actinobacteria/chemistry , Actinobacteria/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/ultrastructure , Adenosine Triphosphate/metabolism , Models, Molecular , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/ultrastructure , Protein Domains , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Structure-Activity Relationship , Thermomonospora , Type VII Secretion Systems/isolation & purification
13.
Cell Stress Chaperones ; 24(6): 1055-1065, 2019 11.
Article En | MEDLINE | ID: mdl-31410727

The eukaryotic cytosolic chaperonin, t-complex polypeptide 1 (TCP-1) ring complex or TRiC, is responsible for folding a tenth of the proteins in the cell. TRiC is a double-ringed barrel with each ring composed of eight different CCT (chaperonin containing TCP-1) subunits. In order for the subunits to assemble together into mature TRiC, which is believed to contain one and only one of each of these subunits per ring, they must be translated from different chromosomes, correctly folded and assembled. When expressed alone in Escherichia coli, the subunits CCT4 and CCT5, interestingly, form TRiC-like homo-oligomeric rings. To explore potential subunit-subunit interactions, we co-expressed these homo-oligomerizing CCT4 and CCT5 subunits or the archaeal chaperonin Mm-Cpn (Methanococcus maripaludis chaperonin) with CCT1-8, one at a time. We found that CCT5 shifted all of the CCT subunits, with the exception of CCT6, into double-barrel TRiC-like complexes, while CCT4 only interacted with CCT5 and CCT8 to form chaperonin rings. We hypothesize that these specific interactions may be due to the formation of hetero-oligomers in E. coli, although more work is needed for validation. We also observed the interaction of CCT5 and Mm-Cpn with smaller fragments of the CCT subunits, confirming their intrinsic chaperone activity. Based on this hetero-oligomer data, we propose that TRiC assembly relies on subunit exchange with some stable homo-oligomers, possibly CCT5, as base assembly units. Eventually, analysis of CCT arrangement in various tissues and at different developmental times is anticipated to provide additional insight on TRiC assembly and CCT subunit composition.


Chaperonin Containing TCP-1 , Cloning, Molecular/methods , Escherichia coli/genetics , Protein Folding , Protein Multimerization/physiology , Protein Subunits , Chaperonin Containing TCP-1/chemistry , Chaperonin Containing TCP-1/isolation & purification , Protein Subunits/chemistry , Protein Subunits/isolation & purification
14.
Proc Natl Acad Sci U S A ; 116(30): 15007-15012, 2019 07 23.
Article En | MEDLINE | ID: mdl-31292253

High-resolution structural information is essential to understand protein function. Protein-structure determination needs a considerable amount of protein, which can be challenging to produce, often involving harsh and lengthy procedures. In contrast, the several thousand to a few million protein particles required for structure determination by cryogenic electron microscopy (cryo-EM) can be provided by miniaturized systems. Here, we present a microfluidic method for the rapid isolation of a target protein and its direct preparation for cryo-EM. Less than 1 µL of cell lysate is required as starting material to solve the atomic structure of the untagged, endogenous human 20S proteasome. Our work paves the way for high-throughput structure determination of proteins from minimal amounts of cell lysate and opens more opportunities for the isolation of sensitive, endogenous protein complexes.


Cryoelectron Microscopy/methods , Image Processing, Computer-Assisted/statistics & numerical data , Proteasome Endopeptidase Complex/ultrastructure , Protein Subunits/chemistry , Biotinylation , Cryoelectron Microscopy/instrumentation , HeLa Cells , Humans , Imaging, Three-Dimensional , Immunoglobulin Fab Fragments/chemistry , Microfluidic Analytical Techniques/methods , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/isolation & purification , Protein Conformation , Protein Subunits/isolation & purification , Vitrification
15.
Protein Expr Purif ; 160: 45-55, 2019 08.
Article En | MEDLINE | ID: mdl-30954531

Packaging the viral genome in the head of double-stranded DNA viruses, such as bacteriophages, requires the activity of a terminase. The bacteriophage terminase consists of a small terminase subunit (TerS), which binds the viral DNA, and a large terminase subunit (TerL) that possesses the ATPase and nuclease activities for packaging the DNA in the phage head. Some phages require additional components for DNA packaging, such as the HNH endonuclease gp74 in the bacteriophage HK97. Gp74 enhances the activity of terminase-mediated digestion of the cohesive (cos) site that connects individual genomes in phage concatemeric DNA, a pre-requisite to DNA packaging, and this enhancement requires an intact HNH motif in gp74. Testing of whether gp74 alters the terminase DNA binding or enzymatic activities requires obtaining isolated samples of pure TerS and TerL, which has been challenging owing to the poor solubility of these proteins. To this end, we developed methods to obtain purified TerS and TerL proteins that are active. TerS is expressed solubly in E. coli as a fusion with SUMO, which can be removed during purification to yield a TerS nonamer (TerS9). Homogenous samples of a TerL monomer are also obtained, but the homogeneity of the sample depends on the solution conditions, as seen for other terminases. DNA binding, ATPase, and nuclease assays demonstrate that our preparations of TerS9 and TerL are functional, and that they also function with gp74. Purified TerS9 and TerL enable studies into the molecular basis by which gp74 regulates terminase activity in phage maturation.


Bacteriophages/enzymology , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/isolation & purification , Viral Proteins/chemistry , Viral Proteins/isolation & purification , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Bacteriophages/chemistry , Bacteriophages/genetics , Bacteriophages/physiology , DNA Packaging , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/virology , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly
16.
Protein Sci ; 28(6): 1071-1082, 2019 06.
Article En | MEDLINE | ID: mdl-30942928

The multi-subunit Ca2+ /calmodulin-dependent protein kinase II (CaMKII) holoenzyme plays a critical role in animal learning and memory. The kinase domain of CaMKII is connected by a flexible linker to a C-terminal hub domain that assembles into a 12- or 14-subunit scaffold that displays the kinase domains around it. Studies on CaMKII suggest that the stoichiometry and dynamic assembly/disassembly of hub oligomers may be important for CaMKII regulation. Although CaMKII is a metazoan protein, genes encoding predicted CaMKII-like hub domains, without associated kinase domains, are found in the genomes of some green plants and bacteria. We show that the hub domains encoded by three related green algae, Chlamydomonas reinhardtii, Volvox carteri f. nagarensis, and Gonium pectoral, assemble into 16-, 18-, and 20-subunit oligomers, as assayed by native protein mass spectrometry. These are the largest known CaMKII hub domain assemblies. A crystal structure of the hub domain from C. reinhardtii reveals an 18-subunit organization. We identified four intra-subunit hydrogen bonds in the core of the fold that are present in the Chlamydomonas hub domain, but not in metazoan hubs. When six point mutations designed to recapitulate these hydrogen bonds were introduced into the human CaMKII-α hub domain, the mutant protein formed assemblies with 14 and 16 subunits, instead of the normal 12- and 14-subunit assemblies. Our results show that the stoichiometric balance of CaMKII hub assemblies can be shifted readily by small changes in sequence.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinase Type 2/isolation & purification , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Domains , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Sequence Alignment
17.
FEBS Lett ; 593(6): 611-621, 2019 03.
Article En | MEDLINE | ID: mdl-30815863

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.


Arabidopsis Proteins/genetics , Arabidopsis/enzymology , Cloning, Molecular/methods , Group I Chaperonins/genetics , Molecular Chaperones/genetics , Phosphate-Binding Proteins/genetics , Ribulose-Bisphosphate Carboxylase/genetics , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Carbon Dioxide/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Group I Chaperonins/metabolism , Histidine/genetics , Histidine/metabolism , Kinetics , Models, Molecular , Molecular Chaperones/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Phosphate-Binding Proteins/metabolism , Photosynthesis/genetics , Protein Folding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Ribulose-Bisphosphate Carboxylase/isolation & purification , Ribulose-Bisphosphate Carboxylase/metabolism
18.
Methods Enzymol ; 619: 179-223, 2019.
Article En | MEDLINE | ID: mdl-30910021

The 20S proteasome is a large multisubunit proteolytic machine that is central to intracellular protein degradation. It is found in all three kingdoms of life and is ubiquitous in archaea and eukaryotes. Since its discovery, much effort employing a diverse array of structural biology methods has been applied to help understand its structure/function relationships. Here, we will specifically focus on the application of native mass spectrometry (MS) approaches for structural investigations of the 20S proteasome. Native MS is a method that examines intact protein assemblies, without disturbing the noncovalent interactions that govern the overall structure. This method is ideally suited to revealing the intrinsic heterogeneity of a given sample and provides insight into the composition, stoichiometry, subunit architecture, and topology of the protein assembly. Initially, we describe native MS-oriented protocols for the isolation of endogenous 20S proteasomes from yeast, rat liver, and human cells. We then highlight the applicability of native MS methodologies, using different instrumental platforms, for structural investigations of the complex. In particular, by means of proteasome biology, we highlight the different approaches used to analyze both intact complexes-their natural heterogeneity and interactions with substrates and regulators-and their individual constituent subunits.


Mass Spectrometry/methods , Proteasome Endopeptidase Complex/chemistry , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Liver/enzymology , Proteasome Endopeptidase Complex/isolation & purification , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Rats
19.
J Bacteriol ; 201(7)2019 04 01.
Article En | MEDLINE | ID: mdl-30642991

In this study, the ATP synthase of Ignicoccus hospitalis was purified, characterized, and structurally compared to the respective enzymes of the other Ignicoccus species, to shed light on energy conservation in this unique group of archaea. The crenarchaeal genus Ignicoccus comprises three described species, i.e., I. hospitalis and Ignicoccus islandicus from hot marine sediments near Iceland and Ignicoccus pacificus from a hydrothermal vent system in the Pacific Ocean. This genus is unique among all archaea due to the unusual cell envelope, consisting of two membranes that enclose a large intermembrane compartment (IMC). I. hospitalis is the best studied member of this genus, mainly because it is the only known host for the potentially parasitic archaeon Nanoarchaeum equitansI. hospitalis grows chemolithoautotrophically, and its sole energy-yielding reaction is the reduction of elemental sulfur with molecular hydrogen, forming large amounts of hydrogen sulfide. This reaction generates an electrochemical gradient, which is used by the ATP synthase, located in the outer cellular membrane, to generate ATP inside the IMC. The genome of I. hospitalis encodes nine subunits of an A-type ATP synthase, which we could identify in the purified complex. Although the maximal in vitro activity of the I. hospitalis enzyme was measured around pH 6, the optimal stability of the A1AO complex seemed to be at pH 9. Interestingly, the soluble A1 subcomplexes of the different Ignicoccus species exhibited significant differences in their apparent molecular masses in native electrophoresis, although their behaviors in gel filtration and chromatography-mass spectrometry were very similar.IMPORTANCE The Crenarchaeota represent one of the major phyla within the Archaea domain. This study describes the successful purification of a crenarchaeal ATP synthase. To date, all information about A-type ATP synthases is from euryarchaeal enzymes. The fact that it has not been possible to purify this enzyme complex from a member of the Crenarchaeota until now points to significant differences in stability, possibly caused by structural alterations. Furthermore, the study subject I. hospitalis has a particular importance among crenarchaeotes, since it is the only known host of N. equitans The energy metabolism in this system is still poorly understood, and our results can help elucidate the unique relationship between these two microbes.


ATP Synthetase Complexes/isolation & purification , ATP Synthetase Complexes/metabolism , Desulfurococcaceae/enzymology , ATP Synthetase Complexes/chemistry , Desulfurococcaceae/isolation & purification , Enzyme Stability , Geologic Sediments , Hydrogen-Ion Concentration , Kinetics , Molecular Weight , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Protein Subunits/metabolism
20.
Nucleic Acids Res ; 47(1): 450-467, 2019 01 10.
Article En | MEDLINE | ID: mdl-30395313

BbvCI, a Type IIT restriction endonuclease, recognizes and cleaves the seven base pair sequence 5'-CCTCAGC-3', generating 3-base, 5'-overhangs. BbvCI is composed of two protein subunits, each containing one catalytic site. Either site can be inactivated by mutation resulting in enzyme variants that nick DNA in a strand-specific manner. Here we demonstrate that the holoenzyme is labile, with the R1 subunit dissociating at low pH. Crystallization of the R2 subunit under such conditions revealed an elongated dimer with the two catalytic sites located on opposite sides. Subsequent crystallization at physiological pH revealed a tetramer comprising two copies of each subunit, with a pair of deep clefts each containing two catalytic sites appropriately positioned and oriented for DNA cleavage. This domain organization was further validated with single-chain protein constructs in which the two enzyme subunits were tethered via peptide linkers of variable length. We were unable to crystallize a DNA-bound complex; however, structural similarity to previously crystallized restriction endonucleases facilitated creation of an energy-minimized model bound to DNA, and identification of candidate residues responsible for target recognition. Mutation of residues predicted to recognize the central C:G base pair resulted in an altered enzyme that recognizes and cleaves CCTNAGC (N = any base).


DNA Cleavage , DNA Restriction Enzymes/chemistry , Holoenzymes/chemistry , Protein Subunits/chemistry , Amino Acid Sequence , Base Sequence , Binding Sites , Catalytic Domain , DNA Restriction Enzymes/genetics , DNA Restriction Enzymes/isolation & purification , Escherichia coli/enzymology , Holoenzymes/genetics , Holoenzymes/isolation & purification , Mutation , Peptides/chemistry , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/isolation & purification
...