Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Photosynth Res ; 155(3): 219-245, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36542271

ABSTRACT

Quantifying cellular components is a basic and important step for understanding how a cell works, how it responds to environmental changes, and for re-engineering cells to produce valuable metabolites and increased biomass. We quantified proteins in the model cyanobacterium Synechocystis sp. PCC 6803 given the general importance of cyanobacteria for global photosynthesis, for synthetic biology and biotechnology research, and their ancestral relationship to the chloroplasts of plants. Four mass spectrometry methods were used to quantify cellular components involved in the biosynthesis of chlorophyll, carotenoid and bilin pigments, membrane assembly, the light reactions of photosynthesis, fixation of carbon dioxide and nitrogen, and hydrogen and sulfur metabolism. Components of biosynthetic pathways, such as those for chlorophyll or for photosystem II assembly, range between 1000 and 10,000 copies per cell, but can be tenfold higher for CO2 fixation enzymes. The most abundant subunits are those for photosystem I, with around 100,000 copies per cell, approximately 2 to fivefold higher than for photosystem II and ATP synthase, and 5-20 fold more than for the cytochrome b6f complex. Disparities between numbers of pathway enzymes, between components of electron transfer chains, and between subunits within complexes indicate possible control points for biosynthetic processes, bioenergetic reactions and for the assembly of multisubunit complexes.


Subject(s)
Synechocystis , Synechocystis/metabolism , Photosystem II Protein Complex/metabolism , Cytochrome b6f Complex/metabolism , Photosynthesis , Chlorophyll/metabolism , Photosystem I Protein Complex/metabolism , Electron Transport
2.
Nat Plants ; 6(12): 1491-1502, 2020 12.
Article in English | MEDLINE | ID: mdl-33257858

ABSTRACT

The insertion of magnesium into protoporphyrin initiates the biosynthesis of chlorophyll, the pigment that underpins photosynthesis. This reaction, catalysed by the magnesium chelatase complex, couples ATP hydrolysis by a ChlID motor complex to chelation within the ChlH subunit. We probed the structure and catalytic function of ChlH using a combination of X-ray crystallography, computational modelling, mutagenesis and enzymology. Two linked domains of ChlH in an initially open conformation of ChlH bind protoporphyrin IX, and the rearrangement of several loops envelops this substrate, forming an active site cavity. This induced fit brings an essential glutamate (E660), proposed to be the key catalytic residue for magnesium insertion, into proximity with the porphyrin. A buried solvent channel adjacent to E660 connects the exterior bulk solvent to the active site, forming a possible conduit for the delivery of magnesium or abstraction of protons.


Subject(s)
Chlorophyll/biosynthesis , Enzyme Activation , Lyases/metabolism , Photosynthesis/physiology , Protoporphyrins/metabolism , Thermosynechococcus/metabolism
3.
Biochem J ; 476(15): 2173-2190, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31320503

ABSTRACT

The reversible docking of small, diffusible redox proteins onto a membrane protein complex is a common feature of bacterial, mitochondrial and photosynthetic electron transfer (ET) chains. Spectroscopic studies of ensembles of such redox partners have been used to determine ET rates and dissociation constants. Here, we report a single-molecule analysis of the forces that stabilise transient ET complexes. We examined the interaction of two components of bacterial photosynthesis, cytochrome c2 and the reaction centre (RC) complex, using dynamic force spectroscopy and PeakForce quantitative nanomechanical imaging. RC-LH1-PufX complexes, attached to silicon nitride AFM probes and maintained in a photo-oxidised state, were lowered onto a silicon oxide substrate bearing dispersed, immobilised and reduced cytochrome c2 molecules. Microscale patterns of cytochrome c2 and the cyan fluorescent protein were used to validate the specificity of recognition between tip-attached RCs and surface-tethered cytochrome c2 Following the transient association of photo-oxidised RC and reduced cytochrome c2 molecules, retraction of the RC-functionalised probe met with resistance, and forces between 112 and 887 pN were required to disrupt the post-ET RC-c2 complex, depending on the retraction velocities used. If tip-attached RCs were reduced instead, the probability of interaction with reduced cytochrome c2 molecules decreased 5-fold. Thus, the redox states of the cytochrome c2 haem cofactor and RC 'special pair' bacteriochlorophyll dimer are important for establishing a productive ET complex. The millisecond persistence of the post-ET cytochrome c2[oxidised]-RC[reduced] 'product' state is compatible with rates of cyclic photosynthetic ET, at physiologically relevant light intensities.


Subject(s)
Bacterial Proteins/metabolism , Cytochromes c/metabolism , Light , Photosynthesis , Rhodobacter sphaeroides/enzymology , Bacterial Proteins/chemistry , Cytochromes c/chemistry , Oxidation-Reduction
4.
Biochem J ; 476(13): 1875-1887, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31164400

ABSTRACT

Magnesium chelatase initiates chlorophyll biosynthesis, catalysing the MgATP2--dependent insertion of a Mg2+ ion into protoporphyrin IX. The catalytic core of this large enzyme complex consists of three subunits: Bch/ChlI, Bch/ChlD and Bch/ChlH (in bacteriochlorophyll and chlorophyll producing species, respectively). The D and I subunits are members of the AAA+ (ATPases associated with various cellular activities) superfamily of enzymes, and they form a complex that binds to H, the site of metal ion insertion. In order to investigate the physical coupling between ChlID and ChlH in vivo and in vitro, ChlD was FLAG-tagged in the cyanobacterium Synechocystis sp. PCC 6803 and co-immunoprecipitation experiments showed interactions with both ChlI and ChlH. Co-production of recombinant ChlD and ChlH in Escherichia coli yielded a ChlDH complex. Quantitative analysis using microscale thermophoresis showed magnesium-dependent binding (Kd 331 ± 58 nM) between ChlD and H. The physical basis for a ChlD-H interaction was investigated using chemical cross-linking coupled with mass spectrometry (XL-MS), together with modifications that either truncate ChlD or modify single residues. We found that the C-terminal integrin I domain of ChlD governs association with ChlH, the Mg2+ dependence of which also mediates the cooperative response of the Synechocystis chelatase to magnesium. The interaction site between the AAA+ motor and the chelatase domain of magnesium chelatase will be essential for understanding how free energy from the hydrolysis of ATP on the AAA+ ChlI subunit is transmitted via the bridging subunit ChlD to the active site on ChlH.


Subject(s)
Lyases/chemistry , Magnesium/chemistry , Recombinant Proteins/chemistry , Synechocystis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Lyases/genetics , Protein Domains , Recombinant Proteins/genetics , Synechocystis/genetics
5.
J Biol Chem ; 293(18): 6672-6681, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29559557

ABSTRACT

Protein transport across the cytoplasmic membrane of bacterial cells is mediated by either the general secretion (Sec) system or the twin-arginine translocase (Tat). The Tat machinery exports folded and cofactor-containing proteins from the cytoplasm to the periplasm by using the transmembrane proton motive force as a source of energy. The Tat apparatus apparently senses the folded state of its protein substrates, a quality-control mechanism that prevents premature export of nascent unfolded or misfolded polypeptides, but its mechanistic basis has not yet been determined. Here, we investigated the innate ability of the model Escherichia coli Tat system to recognize and translocate de novo-designed protein substrates with experimentally determined differences in the extent of folding. Water-soluble, four-helix bundle maquette proteins were engineered to bind two, one, or no heme b cofactors, resulting in a concomitant reduction in the extent of their folding, assessed with temperature-dependent CD spectroscopy and one-dimensional 1H NMR spectroscopy. Fusion of the archetypal N-terminal Tat signal peptide of the E. coli trimethylamine-N-oxide (TMAO) reductase (TorA) to the N terminus of the protein maquettes was sufficient for the Tat system to recognize them as substrates. The clear correlation between the level of Tat-dependent export and the degree of heme b-induced folding of the maquette protein suggested that the membrane-bound Tat machinery can sense the extent of folding and conformational flexibility of its substrates. We propose that these artificial proteins are ideal substrates for future investigations of the Tat system's quality-control mechanism.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Hemeproteins/metabolism , Membrane Transport Proteins/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Circular Dichroism , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Heme-Binding Proteins , Hemeproteins/chemistry , Membrane Transport Proteins/chemistry , Methylamines/metabolism , Models, Molecular , Oxidoreductases, N-Demethylating/metabolism , Periplasm/metabolism , Protein Folding , Protein Sorting Signals , Protein Stability , Protein Transport , Proton Magnetic Resonance Spectroscopy , Substrate Specificity , Temperature
6.
Sci Adv ; 4(1): eaaq1407, 2018 01.
Article in English | MEDLINE | ID: mdl-29387799

ABSTRACT

Chlorophylls are essential cofactors for photosynthesis, which sustains global food chains and oxygen production. Billions of tons of chlorophylls are synthesized annually, yet full understanding of chlorophyll biosynthesis has been hindered by the lack of characterization of the Mg-protoporphyrin IX monomethyl ester oxidative cyclase step, which confers the distinctive green color of these pigments. We demonstrate cyclase activity using heterologously expressed enzyme. Next, we assemble a genetic module that encodes the complete chlorophyll biosynthetic pathway and show that it functions in Escherichia coli. Expression of 12 genes converts endogenous protoporphyrin IX into chlorophyll a, turning E. coli cells green. Our results delineate a minimum set of enzymes required to make chlorophyll and establish a platform for engineering photosynthesis in a heterotrophic model organism.


Subject(s)
Biosynthetic Pathways , Escherichia coli , Metabolic Engineering , Protoporphyrins , Escherichia coli/enzymology , Escherichia coli/genetics , Protoporphyrins/biosynthesis , Protoporphyrins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
7.
Sci Rep ; 7(1): 16807, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196704

ABSTRACT

Techniques such as Stochastic Optical Reconstruction Microscopy (STORM) and Structured Illumination Microscopy (SIM) have increased the achievable resolution of optical imaging, but few fluorescent proteins are suitable for super-resolution microscopy, particularly in the far-red and near-infrared emission range. Here we demonstrate the applicability of CpcA, a subunit of the photosynthetic antenna complex in cyanobacteria, for STORM and SIM imaging. The periodicity and width of fabricated nanoarrays of CpcA, with a covalently attached phycoerythrobilin (PEB) or phycocyanobilin (PCB) chromophore, matched the lines in reconstructed STORM images. SIM and STORM reconstructions of Escherichia coli cells harbouring CpcA-labelled cytochrome bd 1 ubiquinol oxidase in the cytoplasmic membrane show that CpcA-PEB and CpcA-PCB are suitable for super-resolution imaging in vivo. The stability, ease of production, small size and brightness of CpcA-PEB and CpcA-PCB demonstrate the potential of this largely unexplored protein family as novel probes for super-resolution microscopy.


Subject(s)
Phycobilins/metabolism , Phycocyanin/metabolism , Phycoerythrin/metabolism , Synechocystis/metabolism , Bacterial Proteins/metabolism , Light-Harvesting Protein Complexes/chemistry , Light-Harvesting Protein Complexes/metabolism , Photosynthesis , Phycocyanin/chemistry , Stochastic Processes
8.
Nat Commun ; 8(1): 1746, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170493

ABSTRACT

Inorganic phosphate is the major bioavailable form of the essential nutrient phosphorus. However, the concentration of phosphate in most natural habitats is low enough to limit microbial growth. Under phosphate-depleted conditions some bacteria utilise phosphite and hypophosphite as alternative sources of phosphorus, but the molecular basis of reduced phosphorus acquisition from the environment is not fully understood. Here, we present crystal structures and ligand binding affinities of periplasmic binding proteins from bacterial phosphite and hypophosphite ATP-binding cassette transporters. We reveal that phosphite and hypophosphite specificity results from a combination of steric selection and the presence of a P-H…π interaction between the ligand and a conserved aromatic residue in the ligand-binding pocket. The characterisation of high affinity and specific transporters has implications for the marine phosphorus redox cycle, and might aid the use of phosphite as an alternative phosphorus source in biotechnological, industrial and agricultural applications.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Phosphinic Acids/metabolism , Phosphites/metabolism , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Kinetics , Ligands , Models, Molecular , Phylogeny , Prochlorococcus/genetics , Prochlorococcus/metabolism , Pseudomonas stutzeri/genetics , Pseudomonas stutzeri/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Trichodesmium/genetics , Trichodesmium/metabolism
9.
J Am Chem Soc ; 138(20): 6591-7, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27133226

ABSTRACT

In chlorophyll biosynthesis, the magnesium chelatase enzyme complex catalyzes the insertion of a Mg(2+) ion into protoporphyrin IX. Prior to this event, two of the three subunits, the AAA(+) proteins ChlI and ChlD, form a ChlID-MgATP complex. We used microscale thermophoresis to directly determine dissociation constants for the I-D subunits from Synechocystis, and to show that the formation of a ChlID-MgADP complex, mediated by the arginine finger and the sensor II domain on ChlD, is necessary for the assembly of the catalytically active ChlHID-MgATP complex. The N-terminal AAA(+) domain of ChlD is essential for complex formation, but some stability is preserved in the absence of the C-terminal integrin domain of ChlD, particularly if the intervening polyproline linker region is retained. Single molecule force spectroscopy (SMFS) was used to determine the factors that stabilize formation of the ChlID-MgADP complex at the single molecule level; ChlD was attached to an atomic force microscope (AFM) probe in two different orientations, and the ChlI subunits were tethered to a silica surface; the probability of subunits interacting more than doubled in the presence of MgADP, and we show that the N-terminal AAA(+) domain of ChlD mediates this process, in agreement with the microscale thermophoresis data. Analysis of the unbinding data revealed a most probable interaction force of around 109 pN for formation of single ChlID-MgADP complexes. These experiments provide a quantitative basis for understanding the assembly and function of the Mg chelatase complex.


Subject(s)
Bacterial Proteins/chemistry , Lyases/chemistry , Nucleotides/chemistry , Catalysis , Synechocystis/chemistry , Synechocystis/enzymology
10.
FEBS Lett ; 590(12): 1687-93, 2016 06.
Article in English | MEDLINE | ID: mdl-27176620

ABSTRACT

In the first committed reaction of chlorophyll biosynthesis, magnesium chelatase couples ATP hydrolysis to the thermodynamically unfavorable Mg(2+) insertion into protoporphyrin IX (ΔG°' of circa 25-33 kJ·mol(-1) ). We explored the thermodynamic constraints on magnesium chelatase and demonstrate the effect of nucleotide hydrolysis on both the reaction kinetics and thermodynamics. The enzyme produces a significant rate enhancement (kcat /kuncat of 400 × 10(6) m) and a catalytic rate enhancement, kcat/KmDIXK0.5Mgkuncat, of 30 × 10(15) m(-1) , increasing to 300 × 10(15) m(-1) with the activator protein Gun4. This is the first demonstration of the thermodynamic benefit of ATP hydrolysis in the AAA(+) family.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/chemistry , Lyases/chemistry , Magnesium/chemistry , Protoporphyrins/chemistry , Synechocystis/enzymology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Catalysis , Lyases/metabolism , Magnesium/metabolism , Protoporphyrins/biosynthesis
11.
Mol Microbiol ; 99(2): 307-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26419219

ABSTRACT

The mature architecture of the photosynthetic membrane of the purple phototroph Rhodobacter sphaeroides has been characterised to a level where an atomic-level membrane model is available, but the roles of the putative assembly proteins LhaA and PucC in establishing this architecture are unknown. Here we investigate the assembly of light-harvesting LH2 and reaction centre-light-harvesting1-PufX (RC-LH1-PufX) photosystem complexes using spectroscopy, pull-downs, native gel electrophoresis, quantitative mass spectrometry and fluorescence lifetime microscopy to characterise a series of lhaA and pucC mutants. LhaA and PucC are important for specific assembly of LH1 or LH2 complexes, respectively, but they are not essential; the few LH1 subunits found in ΔlhaA mutants assemble to form normal RC-LH1-PufX core complexes showing that, once initiated, LH1 assembly round the RC is cooperative and proceeds to completion. LhaA and PucC form oligomers at sites of initiation of membrane invagination; LhaA associates with RCs, bacteriochlorophyll synthase (BchG), the protein translocase subunit YajC and the YidC membrane protein insertase. These associations within membrane nanodomains likely maximise interactions between pigments newly arriving from BchG and nascent proteins within the SecYEG-SecDF-YajC-YidC assembly machinery, thereby co-ordinating pigment delivery, the co-translational insertion of LH polypeptides and their folding and assembly to form photosynthetic complexes.


Subject(s)
Bacterial Proteins/metabolism , Light-Harvesting Protein Complexes/metabolism , Photosystem II Protein Complex/metabolism , Rhodobacter sphaeroides/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Light , Light-Harvesting Protein Complexes/genetics , Photosystem II Protein Complex/genetics , Rhodobacter sphaeroides/genetics , Rhodobacter sphaeroides/radiation effects
12.
Biochemistry ; 54(44): 6659-62, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26513685

ABSTRACT

Magnesium chelatase catalyzes the first committed step in chlorophyll biosynthesis by inserting a Mg(2+) ion into protoporphyrin IX in an ATP-dependent manner. The cyanobacterial (Synechocystis) and higher-plant chelatases exhibit a complex cooperative response to free magnesium, while the chelatases from Thermosynechococcus elongatus and photosynthetic bacteria do not. To investigate the basis for this cooperativity, we constructed a series of chimeric ChlD proteins using N-terminal, central, and C-terminal domains from Synechocystis and Thermosynechococcus. We show that five glutamic acid residues in the C-terminal domain play a major role in this process.


Subject(s)
Bacterial Proteins/metabolism , Lyases/metabolism , Magnesium/metabolism , Synechococcus/metabolism , Synechocystis/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Cations, Divalent/metabolism , Glutamic Acid/analysis , Glutamic Acid/metabolism , Lyases/chemistry , Molecular Sequence Data , Protein Structure, Tertiary , Synechococcus/chemistry , Synechocystis/chemistry
13.
J Biol Chem ; 290(47): 28477-28488, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26446792

ABSTRACT

In oxygenic phototrophs, chlorophylls, hemes, and bilins are synthesized by a common branched pathway. Given the phototoxic nature of tetrapyrroles, this pathway must be tightly regulated, and an important regulatory role is attributed to magnesium chelatase enzyme at the branching between the heme and chlorophyll pathway. Gun4 is a porphyrin-binding protein known to stimulate in vitro the magnesium chelatase activity, but how the Gun4-porphyrin complex acts in the cell was unknown. To address this issue, we first performed simulations to determine the porphyrin-docking mechanism to the cyanobacterial Gun4 structure. After correcting crystallographic loop contacts, we determined the binding site for magnesium protoporphyrin IX. Molecular modeling revealed that the orientation of α6/α7 loop is critical for the binding, and the magnesium ion held within the porphyrin is coordinated by Asn-211 residue. We also identified the basis for stronger binding in the Gun4-1 variant and for weaker binding in the W192A mutant. The W192A-Gun4 was further characterized in magnesium chelatase assay showing that tight porphyrin binding in Gun4 facilitates its interaction with the magnesium chelatase ChlH subunit. Finally, we introduced the W192A mutation into cells and show that the Gun4-porphyrin complex is important for the accumulation of ChlH and for channeling metabolites into the chlorophyll biosynthetic pathway.


Subject(s)
Bacterial Proteins/metabolism , Chlorophyll/biosynthesis , Porphyrins/metabolism , Synechocystis/metabolism , Bacterial Proteins/chemistry , Circular Dichroism , Crystallography, X-Ray , Molecular Dynamics Simulation , Mutation , Protein Conformation , Synechocystis/genetics , Synechocystis/growth & development
14.
Biochem J ; 464(3): 315-22, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25471602

ABSTRACT

Magnesium chelatase (MgCH) initiates chlorophyll biosynthesis by catalysing the ATP-dependent insertion of Mg2+ into protoporphyrin. This large enzyme complex comprises ChlH, I and D subunits, with I and D involved in ATP hydrolysis, and H the protein that handles the substrate and product. The 148 kDa ChlH subunit has a globular N-terminal domain attached by a narrow linker to a hollow cage-like structure. Following deletion of this ~18 kDa domain from the Thermosynechoccus elongatus ChlH, we used single particle reconstruction to show that the apo- and porphyrin-bound forms of the mutant subunit consist of a hollow globular protein with three connected lobes; superposition of the mutant and native ChlH structures shows that, despite the clear absence of the N-terminal 'head' region, the rest of the protein appears to be correctly folded. Analyses of dissociation constants shows that the ΔN159ChlH mutant retains the ability to bind protoporphyrin and the Gun4 enhancer protein, although the addition of I and D subunits yields an extremely impaired active enzyme complex. Addition of the Gun4 enhancer protein, which stimulates MgCH activity significantly especially at low Mg2+ concentrations, partially reactivates the ΔN159ChlH-I-D mutant enzyme complex, suggesting that the binding site or sites for Gun4 on H do not wholly depend on the N-terminal domain.


Subject(s)
Lyases/chemistry , Lyases/physiology , Synechococcus/enzymology , Amino Acid Sequence , Gene Deletion , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship
15.
Biochim Biophys Acta ; 1837(10): 1769-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24530865

ABSTRACT

Photosynthesis converts absorbed solar energy to a protonmotive force, which drives ATP synthesis. The membrane network of chlorophyll-protein complexes responsible for light absorption, photochemistry and quinol (QH2) production has been mapped in the purple phototrophic bacterium Rhodobacter (Rba.) sphaeroides using atomic force microscopy (AFM), but the membrane location of the cytochrome bc1 (cytbc1) complexes that oxidise QH2 to quinone (Q) to generate a protonmotive force is unknown. We labelled cytbc1 complexes with gold nanobeads, each attached by a Histidine10 (His10)-tag to the C-terminus of cytc1. Electron microscopy (EM) of negatively stained chromatophore vesicles showed that the majority of the cytbc1 complexes occur as dimers in the membrane. The cytbc1 complexes appeared to be adjacent to reaction centre light-harvesting 1-PufX (RC-LH1-PufX) complexes, consistent with AFM topographs of a gold-labelled membrane. His-tagged cytbc1 complexes were retrieved from chromatophores partially solubilised by detergent; RC-LH1-PufX complexes tended to co-purify with cytbc1 whereas LH2 complexes became detached, consistent with clusters of cytbc1 complexes close to RC-LH1-PufX arrays, but not with a fixed, stoichiometric cytbc1-RC-LH1-PufX supercomplex. This information was combined with a quantitative mass spectrometry (MS) analysis of the RC, cytbc1, ATP synthase, cytaa3 and cytcbb3 membrane protein complexes, to construct an atomic-level model of a chromatophore vesicle comprising 67 LH2 complexes, 11 LH1-RC-PufX dimers & 2 RC-LH1-PufX monomers, 4 cytbc1 dimers and 2 ATP synthases. Simulation of the interconnected energy, electron and proton transfer processes showed a half-maximal ATP turnover rate for a light intensity equivalent to only 1% of bright sunlight. Thus, the photosystem architecture of the chromatophore is optimised for growth at low light intensities.


Subject(s)
Electron Transport , Photosynthesis , Rhodobacter sphaeroides/metabolism , Base Sequence , DNA Primers , Mass Spectrometry , Microscopy, Atomic Force , Models, Molecular , Spectrophotometry, Ultraviolet
16.
Photosynth Res ; 120(1-2): 169-80, 2014 May.
Article in English | MEDLINE | ID: mdl-23539360

ABSTRACT

Electron transfer pathways in photosynthesis involve interactions between membrane-bound complexes such as reaction centres with an extrinsic partner. In this study, the biological specificity of electron transfer between the reaction centre-light-harvesting 1-PufX complex and its extrinsic electron donor, cytochrome c 2, formed the basis for mapping the location of surface-attached RC-LH1-PufX complexes using atomic force microscopy (AFM). This nano-mechanical mapping method used an AFM probe functionalised with cyt c 2 molecules to quantify the interaction forces involved, at the single-molecule level under native conditions. With surface-bound RC-His12-LH1-PufX complexes in the photo-oxidised state, the mean interaction force with cyt c 2 is approximately 480 pN with an interaction frequency of around 66 %. The latter value lowered 5.5-fold when chemically reduced RC-His12-LH1-PufX complexes are imaged in the dark to abolish electron transfer from cyt c 2 to the RC. The correspondence between topographic and adhesion images recorded over the same area of the sample shows that affinity-based AFM methods are a useful tool when topology alone is insufficient for spatially locating proteins at the surface of photosynthetic membranes.


Subject(s)
Cytochromes c2/metabolism , Microscopy, Atomic Force , Photosynthesis/physiology , Electron Transport/physiology , Models, Biological , Rhodobacter sphaeroides/metabolism
17.
Biochem J ; 457(1): 163-70, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24138165

ABSTRACT

The first committed step in chlorophyll biosynthesis is catalysed by magnesium chelatase (E.C. 6.6.1.1), which uses the free energy of ATP hydrolysis to insert an Mg(2+) ion into the ring of protoporphyrin IX. We have characterized magnesium chelatase from the thermophilic cyanobacterium Thermosynechococcus elongatus. This chelatase is thermostable, with subunit melting temperatures between 55 and 63°C and optimal activity at 50°C. The T. elongatus chelatase (kcat of 0.16 µM/min) shows a Michaelis-Menten-type response to both Mg(2+) (Km of 2.3 mM) and MgATP(2-) (Km of 0.8 mM). The response to porphyrin is more complex; porphyrin inhibits at high concentrations of ChlH, but when the concentration of ChlH is comparable with the other two subunits the response is of a Michaelis-Menten type (at 0.4 µM ChlH, Km is 0.2 µM). Hybrid magnesium chelatases containing a mixture of subunits from the mesophilic Synechocystis and Thermosynechococcus enzymes are active. We generated all six possible hybrid magnesium chelatases; the hybrid chelatase containing Thermosynechococcus ChlD and Synechocystis ChlI and ChlH is not co-operative towards Mg(2+), in contrast with the Synechocystis magnesium chelatase. This loss of co-operativity reveals the significant regulatory role of Synechocystis ChlD.


Subject(s)
Cyanobacteria/enzymology , Lyases/physiology , Adenosine Triphosphate/pharmacology , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Enzyme Activation , Kinetics , Lyases/chemistry , Lyases/isolation & purification , Magnesium/pharmacology , Osmolar Concentration , Protein Subunits/physiology , Synechocystis/enzymology , Temperature
18.
Biochemistry ; 52(43): 7575-85, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24131108

ABSTRACT

Reaction center-light harvesting 1 (RC-LH1) complexes are the fundamental units of bacterial photosynthesis, which use solar energy to power the reduction of quinone to quinol prior to the formation of the proton gradient that drives ATP synthesis. The dimeric RC-LH1-PufX complex of Rhodobacter sphaeroides is composed of 64 polypeptides and 128 cofactors, including 56 LH1 bacteriochlorophyll a (BChl a) molecules that surround and donate energy to the two RCs. The 3D structure was determined to 8 Å by X-ray crystallography, and a model was built with constraints provided by electron microscopy (EM), nuclear magnetic resonance (NMR), mass spectrometry (MS), and site-directed mutagenesis. Each half of the dimer complex consists of a RC surrounded by an array of 14 LH1 αß subunits, with two BChls sandwiched between each αß pair of transmembrane helices. The N- and C-terminal extrinsic domains of PufX promote dimerization by interacting with the corresponding domains of an LH1 ß polypeptide from the other half of the RC-LH1-PufX complex. Close contacts between PufX, an LH1 αß subunit, and the cytoplasmic domain of the RC-H subunit prevent the LH1 complex from encircling the RC and create a channel connecting the RC QB site to an opening in the LH1 ring, allowing Q/QH2 exchange with the external quinone pool. We also identified a channel that connects the two halves of the dimer, potentially forming a long-range pathway for quinone migration along rows of RC-LH1-PufX complexes in the membrane. The structure of the RC-LH1-PufX complex explains the crucial role played by PufX in dimer formation, and it shows how quinone traffic traverses the LH1 complex as it shuttles between the RC and the cytochrome bc1 complex.


Subject(s)
Bacterial Proteins/chemistry , Light-Harvesting Protein Complexes/chemistry , Models, Molecular , Rhodobacter sphaeroides/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriochlorophyll A/analysis , Bacteriochlorophyll A/chemistry , Bacteriochlorophyll A/metabolism , Benzoquinones/chemistry , Benzoquinones/metabolism , Carotenoids/analysis , Carotenoids/chemistry , Carotenoids/metabolism , Light-Harvesting Protein Complexes/genetics , Light-Harvesting Protein Complexes/metabolism , Mass Spectrometry , Oxidation-Reduction , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , X-Ray Diffraction
19.
J Biol Chem ; 287(7): 4946-56, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22179610

ABSTRACT

The biosynthesis of chlorophyll, an essential cofactor for photosynthesis, requires the ATP-dependent insertion of Mg(2+) into protoporphyrin IX catalyzed by the multisubunit enzyme magnesium chelatase. This enzyme complex consists of the I subunit, an ATPase that forms a complex with the D subunit, and an H subunit that binds both the protoporphyrin substrate and the magnesium protoporphyrin product. In this study we used electron microscopy and small-angle x-ray scattering to investigate the structure of the magnesium chelatase H subunit, ChlH, from the thermophilic cyanobacterium Thermosynechococcus elongatus. Single particle reconstruction of negatively stained apo-ChlH and Chl-porphyrin proteins was used to reconstitute three-dimensional structures to a resolution of ∼30 Å. ChlH is a large, 148-kDa protein of 1326 residues, forming a cage-like assembly comprising the majority of the structure, attached to a globular N-terminal domain of ∼16 kDa by a narrow linker region. This N-terminal domain is adjacent to a 5 nm-diameter opening in the structure that allows access to a cavity. Small-angle x-ray scattering analysis of ChlH, performed on soluble, catalytically active ChlH, verifies the presence of two domains and their relative sizes. Our results provide a basis for the multiple regulatory and catalytic functions of ChlH of oxygenic photosynthetic organisms and for a chaperoning function that sequesters the enzyme-bound magnesium protoporphyrin product prior to its delivery to the next enzyme in the chlorophyll biosynthetic pathway, magnesium protoporphyrin methyltransferase.


Subject(s)
Bacterial Proteins/chemistry , Cyanobacteria/enzymology , Lyases/chemistry , Models, Molecular , Protein Structure, Tertiary , Protein Subunits/chemistry , Scattering, Small Angle , X-Ray Diffraction
20.
Proc Natl Acad Sci U S A ; 108(1): 97-102, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173279

ABSTRACT

The class II chelatases associated with heme, siroheme, and cobalamin biosynthesis are structurally related enzymes that insert a specific metal ion (Fe(2+) or Co(2+)) into the center of a modified tetrapyrrole (protoporphyrin or sirohydrochlorin). The structures of two related class II enzymes, CbiX(S) from Archaeoglobus fulgidus and CbiK from Salmonella enterica, that are responsible for the insertion of cobalt along the cobalamin biosynthesis pathway are presented in complex with their metallated product. A further structure of a CbiK from Desulfovibrio vulgaris Hildenborough reveals how cobalt is bound at the active site. The crystal structures show that the binding of sirohydrochlorin is distinctly different to porphyrin binding in the protoporphyrin ferrochelatases and provide a molecular overview of the mechanism of chelation. The structures also give insights into the evolution of chelatase form and function. Finally, the structure of a periplasmic form of Desulfovibrio vulgaris Hildenborough CbiK reveals a novel tetrameric arrangement of its subunits that are stabilized by the presence of a heme b cofactor. Whereas retaining colbaltochelatase activity, this protein has acquired a central cavity with the potential to chaperone or transport metals across the periplasmic space, thereby evolving a new use for an ancient protein subunit.


Subject(s)
Cobalt/metabolism , Evolution, Molecular , Ferrochelatase/metabolism , Models, Molecular , Multigene Family/genetics , Vitamin B 12/biosynthesis , Archaeoglobus fulgidus/enzymology , Catalytic Domain/genetics , Crystallization , Desulfovibrio vulgaris/enzymology , Ferrochelatase/genetics , Porphyrins/metabolism , Salmonella enterica/enzymology , Uroporphyrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...