Mechanistic aspects of the Q o -site of the bc 1 -complex as revealed by mutagenesis studies, and the crystallographic structure.

Similar documents
Antony R. Crofts,*, Blanca Barquera, Robert B. Gennis,, Richard Kuras, Mariana Guergova-Kuras, and Edward A. Berry*,

Mechanism of Ubiquinol Oxidation by the bc 1 Complex: Role of the Iron Sulfur Protein and Its Mobility

Evidence for a Concerted Mechanism of Ubiquinol Oxidation by the Cytochrome bc 1 Complex*

Exploration of Ligands to the Q i Site Semiquinone in the bc 1 Complex Using High-resolution EPR*

Electron Transfer Chain

In Situ Kinetics of Cytochromes c 1 and c 2

Modulation of the Midpoint Potential of the [2Fe-2S] Rieske Iron Sulfur Center by Q o Occupants in the bc 1 Complex

The cytochrome bc 1 complex of Rhodobacter capsulatus: ubiquinol oxidation in a dimeric Q-cycle?

MEMBRANE-BOUND ELECTRON TRANSFER AND ATP SYNTHESIS (taken from Chapter 18 of Stryer)

Movement of the Iron-Sulfur Head Domain of Cytochrome bc 1 Transiently Opens the Catalytic Q o Site for Reaction with Oxygen

Anti-cooperative Oxidation of Ubiquinol by the Yeast Cytochrome bc 1 Complex* S

Biochimica et Biophysica Acta

Oxidative Phosphorylation

Photosynthesis: The light Reactions. Dr. Obaidur Rahman NSU

Chapter 14 - Electron Transport and Oxidative Phosphorylation

19 Oxidative Phosphorylation and Photophosphorylation W. H. Freeman and Company

Chemical Mechanism of Enzymes

Porphyrins: Chemistry and Biology

Oxidative phosphorylation & Photophosphorylation

Light-Driven Proton Translocation

Maria B. Valkova-Valchanova, A. Sami Saribas, Brian R. Gibney, P. Leslie Dutton, and Fevzi Daldal*, Biochemistry 1998, 37,

بسم هللا الرحمن الرحيم

P450 CYCLE. All P450s follow the same catalytic cycle of;

Multiple Q-Cycle Bypass Reactions at the Q o Site of the Cytochrome bc 1 Complex

The Q-cycle a personal perspective

BioChem Course Outline

Electron Transport System Supplemental Reading. Key Concepts PETER MITCHELL'S CHEMIOSMOTIC THEORY

Myxothiazol Induces H 2 O 2 Production from Mitochondrial Respiratory Chain

Photo-induced cyclic electron transfer operates in frozen cells of Rhodobacter sphaeroides

Vocabulary. Chapter 20: Electron Transport and Oxidative Phosphorylation

Statin inhibition of HMG-CoA reductase: a 3-dimensional view

SUPPORTING INFORMATION. Lysine Carbonylation is a Previously Unrecognized Contributor. to Peroxidase Activation of Cytochrome c by Chloramine-T

Electron Transport Chain and Oxidative phosphorylation

Chapter 7. Heme proteins Cooperativity Bohr effect

Chapter 11: Enzyme Catalysis

Synthesis of ATP, the energy currency in metabolism

Detergent solubilised 5 TMD binds pregnanolone at the Q245 neurosteroid potentiation site.

Globular proteins Proteins globular fibrous

Amino Acids. Review I: Protein Structure. Amino Acids: Structures. Amino Acids (contd.) Rajan Munshi

Structure of the Yeast Cytochrome bc 1 Complex with a Hydroxyquinone Anion Q o Site Inhibitor Bound*

Tema 4. Electron Transport. Cap. 4 pages

Mechanism of electron transfer in the cytochrome b/f complex of algae: Evidence for a semiquinone cycle

Biochemistry: A Short Course Second Edition

This is the first of two chapters that describe respiration in the mitochondria. The word

Nafith Abu Tarboush DDS, MSc, PhD

A. Sami Sarıbaş, Maria Valkova-Valchanova, Mariko K. Tokito,, Zhaolei Zhang, Edward A. Berry, and Fevzi Daldal*, Biochemistry 1998, 37,

) one consumes in breathing is converted to:, which of the following would be found in the oxidized state?

Electron transport chain, oxidative phosphorylation, mitochondrial transport systems

This exam consists of two parts. Part I is multiple choice. Each of these 25 questions is worth 2 points.

Superoxide anion generation by the cytochrome bc 1 complex. Jian Sun and Bernard L. Trumpower *

3.2 Ligand-Binding at Nicotinic Acid Receptor Subtypes GPR109A/B

2. Which of the following amino acids is most likely to be found on the outer surface of a properly folded protein?

Electrogenic events associated with electron and proton transfers within the cytochrome b 6 /f complex

Received 27 July 1993

Nafith Abu Tarboush DDS, MSc, PhD

Copyright 2010 Doreen C. Victoria All rights reserved

Electron Transport and Oxidative. Phosphorylation

Electron transport chain chapter 6 (page 73) BCH 340 lecture 6

Supplementary Information

Inhibition of Reversed Electron Transfer and Proton Transport in the Beef Heart Cytochrome bc 1 Complex by Chemical Modification

Cbl ubiquitin ligase: Lord of the RINGs

Phenylketonuria (PKU) Structure of Phenylalanine Hydroxylase. Biol 405 Molecular Medicine

Transient β-hairpin Formation in α-synuclein Monomer Revealed by Coarse-grained Molecular Dynamics Simulation

Continued Study of Proton Pumping by Incorporation of a Genetically Encoded Infrared Probe. UROP Assistantship Proposal Fall 2015

Crystallographic location of two Zn 2 -binding sites in the avian cytochrome bc 1 complex

Excerpt from J. Mol. Biol. (2002) 320, :

Biochemistry 15 Doctor /7/2012

Electron Transport and Carbon Fixation in Chloroplasts

Chemistry 5.07 Problem Set 5 (redox cofactors and oxidation reactions; carbohydrate chemistry, and introduction to metabolism)

CELL BIOLOGY - CLUTCH CH AEROBIC RESPIRATION.

PBL SEMINAR. HEMOGLOBIN, O 2 -TRANSPORT and CYANOSIS An Overview

CHAPTER 4. Tryptophan fluorescence quenching by brominated lipids

Crystal Structure of the Subtilisin Carlsberg: OMTKY3 Complex

number Done by Corrected by Doctor Nafeth Abu Tarboush

Part III => METABOLISM and ENERGY. 3.6 Oxidative Phosphorylation 3.6a Electron Transport 3.6b ATP Synthesis

List of Figures. List of Tables

PHAR3316 Pharmacy biochemistry Exam #2 Fall 2010 KEY

The cytochrome bc 1 complex and its homologue the b 6 f complex: similarities and differences

The MOLECULES of LIFE

Catalysis & specificity: Proteins at work

Biochemistry - I SPRING Mondays and Wednesdays 9:30-10:45 AM (MR-1307) Lectures Based on Profs. Kevin Gardner & Reza Khayat

OVERVIEW OF RESPIRATION AND LOOSE ENDS. What agents? What war?

PAPER No. : 16, Bioorganic and biophysical chemistry MODULE No. : 22, Mechanism of enzyme catalyst reaction (I) Chymotrypsin

Supplementary Figure 1 (previous page). EM analysis of full-length GCGR. (a) Exemplary tilt pair images of the GCGR mab23 complex acquired for Random

Chapter VI. Increased affinity in the complex of yeast cytochrome c and cytochrome c peroxidase

Evolution of Photosynthesis: Time-Independent Structure of the Cytochrome b 6 f Complex

Elke Denke, Torsten Merbitz-Zahradnik, Oliver M. Hatzfeld, Christopher H. Snyder, Thomas A. Link, and Bernard L. Trumpower

SUPPORTING INFORMATION FOR. A Computational Approach to Enzyme Design: Using Docking and MM- GBSA Scoring

Supplementary Figure 1 Preparation, crystallization and structure determination of EpEX. (a), Purified EpEX and EpEX analyzed on homogenous 12.

Final Exam Chemistry 391 Structural Biochemistry Fall Do not open the exam until ready to begin! Rules of the Game:

Nature Structural & Molecular Biology: doi: /nsmb Supplementary Figure 1

6. C-type cytochrome, soluble and membrane protein

Torsten Merbitz-Zahradnik, Klaus Zwicker, Jürgen H. Nett, Thomas A. Link, and Bernard L. Trumpower*, Biochemistry 2003, 42,

Key Concepts. Learning Objectives

Multiple-Choice Questions Answer ALL 20 multiple-choice questions on the Scantron Card in PENCIL

Enzymes: The Catalysts of Life

Proteins. Amino acids, structure and function. The Nobel Prize in Chemistry 2012 Robert J. Lefkowitz Brian K. Kobilka

1. Hemoglobin and the Movement of Oxygen. Respirator system/biochemistry

Plant Respiration. Exchange of Gases in Plants:

(B D) Three views of the final refined 2Fo-Fc electron density map of the Vpr (red)-ung2 (green) interacting region, contoured at 1.4σ.

Transcription:

Mechanistic aspects of the Q o -site of the bc 1 -complex as revealed by mutagenesis studies, and the crystallographic structure. By A.R. Crofts*, Blanca Barquera*, R.B. Gennis*, R. Kuras*, Mariana Guergova-Kuras* and E.A. Berry # Center for Biophysics and Computational Biology, U. of Illinois at Urbana-Champaign*, and Lawrence Berkeley National Laboratory, U. C. Berkeley #. Abstract Solution of the X-ray crystallographic structure of the ubiquinol:cytochrome c oxidoreductase (the bc 1 -complex) from several animal sources has provide a basis for understanding the mechanism, and the effects of mutation on the function of the complex. In this paper, we review the biophysical information on the mechanism of the ubihydroquinone (quinol) oxidizing site (the Q o -site), the information from analysis of mutant strains which effect function or inhibitor binding at the quinol oxidizing site, and details of the structure of the complex from avian heart mitochondria as it pertains to this catalytic site. A paradoxical feature of the quinol oxidation reaction is the mechanism that ensures that the two electrons from quinol enter separate electron transfer chains. The crystallographic data show several features of interest. The iron sulfur protein is found in several different positions in different structures, and in structures with inhibitors bound, stigmatellin and myxothiazol occupy different domains in the Q o -binding pocket, with the former distal and the latter proximal to heme b L. We suggest a mechanism with the following novel features: i) movement of the iron sulfur protein between two separate reaction interfaces on cytochrome c 1 and cytochrome b, and ii) movement of the occupant of the Q o -binding pocket between a domain distal from the heme of cyt b L, but close to the iron sulfur protein docking interface, and a domain proximal to heme b L. We suggest that all interactions with the iron sulfur protein occur from the distal position. These include a) the binding of stigmatellin and UHDBT, which lead to a change in mid-point potential and a shift in spectrum of the reduced 2Fe2S center, b) the formation of a complex between ubiquinone and the reduced iron sulfur protein which gives rise to the g x =1.800 EPR band, and c) the oxidation of quinol to ubisemiquinone by the oxidized 2Fe2S center. We suggest that after formation, the ubisemiquinone moves to the proximal position before transferring an electron to heme b L, and that this movement, together with a change in conformation of the site through displacement of a few residues, prevents further interaction with the 2Fe2S center. Introduction Ubiquinol:cytochrome c oxidoreductase (bc 1 -complex) is a central component of the electron transfer system in almost all organisms, occurring ubiquitously in respiratory and photosynthetic chains of mitochondria and bacteria, and (as the b 6 f-complex) in the photosynthetic chain of oxygenic photosynthesis (see refs. 1-3 for recent reviews). A modified Q-cycle (4-9) accounts well for a large body of experimental data from studies in which the function has been explored.

The mechanism involves two catalytic sites for oxidation or reduction of the quinone couple, and a third site for electron transfer to cytochrome c. The function of the two quinone reactive sites has been explored biochemically, and characterized using kinetic spectroscopy, and their activities differentiated through use of inhibitors that act specifically at one site or the other (1-13). The quinol oxidizing site (Q o -site) catalyzes a unique reaction in which the electrons from QH 2 are passed to separate electron transport chains in the complex through a bifurcated reaction. Further information on the occupancy of the Q o -site has been obtained though study of the interaction of the occupant with the 2Fe2S center of the reduced ISP, observed through changes in the EPR spectrum of this center (33-40). In addition to the effects observed with stigmatellin and UHDBT (10-13, 33), a well-defined band at g x =1.800 has been observed when quinone occupies the pocket. The dependence of these spectral changes on ambient redox potential, and on the extent of extraction of quinone, has lead Ding (34, 35) and colleagues to suggest a double occupancy of the site by weakly (Q ow ) and strongly (Q os ) binding quinone species. Although the modified Q-cycle mechanism is well supported, there remain several problems in understanding the mechanism of catalysis at the molecular level. A more detailed understanding of the structure-function relationship at the catalytic sites requires a structural context, and this has recently been provided by X-ray crystallographic studies of the structure of the complexes from mitochondria (15-23). In this paper, we review the information on mechanism obtained from studies of mutations that effect the Q o -site, and consider this in the context of the detailed structural information from crystallographic studies of the chicken heart mitochondrial bc 1 - complex (20-22). Methods The crystallographic determination of the structures of the complexes is discussed at length elsewhere (22). Molecular engineering protocols used were similar to those previously reported (24-26), and will be presented in detail elsewhere (Barquera et al., in preparation). Biophysical characterization of mutant strains was performed using procedures previously described (5, 6, 9, 11). Results and Discussion Structure of the catalytic subunits The catalytic subunits, cytochrome b, cytochrome c 1 and the iron sulfur protein, have been well conserved across the bacterial/eukaryote evolutionary divide, allowing us to interpret data from experiments with Rb. sphaeroides in the context of the mitochondrial structures. The X-ray crystallographic work has confirmed the general features anticipated in earlier work (1, 29, 30), and provided a wealth of additional detail (22, 23). The location of the quinone reactive sites in the avian structure (22) has been determined using inhibitors shown through functional studies to be specific for each site. Crystals have been grown in the presence of three Q o -site inhibitors, stigmatellin, myxothiazol, and UHDBT, and the Q i -site inhibitor antimycin. The position of the inhibitors has been determined from the electron density difference from the native crystals. The structure of the protein has not yet been determined independently for most of the inhibitor containing crystals, but, apart from changes in a few key residues (to be described in context), the

cytochrome b subunit does not appear to undergo any major changes in structure on binding inhibitors. Figure 1. Scheme showing the reactions of quinol oxidation at the Q o -site. The complex is represented by the three catalytic subunits (cyt b - red; cyt c 1 - green; ISP - blue), shown as transparent spacefilling models, revealing the metal centers. The transfer of an oxidizing equivalent to cyt c 1 is shown by the ringed "+" (a), which is transferred to the ISP (in ISP R position) (b); the oxidation of QH 2 (a-c) is indicated by the appearance of an electron (ringed "-") at the Q o -site, indicating formation of a semiquinone (d) (in c and d, ISP in ISP O position), and its transfer to the b-cytochrome chain (e, f), leaving quinone (e), which leaves the site (f). Note that the electron transfer from QH 2 to the 2Fe2S center to form the semiquinone anion, releases 2H + to the aqueous phase (d), balancing the charges in the transition (c - d). In contrast, the ISP changes its position dramatically on binding stigmatellin, showing a rotational displacement in which the 2Fe2S center moves by ~16 Å from a position close to cyt c 1 in the absence, to a docking interface on cyt b in the presence of inhibitor. Theoretical considerations suggest that the structure would not be competent in all electron transfer steps in either of the positions found, and we have suggested that the ISP must move between these positions during turn over. Figure 1 summarizes the mechanism we have proposed, in which the movement of the catalytic domain of the ISP between reaction interfaces on cytochrome b and cytochrome c 1 subunits is required for electron transfer from quinol to cyt c 1 (31).

The Q o -site An extensive literature on mutations that affect inhibitor binding or kinetic function at the Q o -site has been summarized recently by Brasseur et al. (27); our own recent work on site-directed mutations in the ef-loop, including the -PEWY- span, will be presented in detail elsewhere (28, and Barquera et al., in preparation). This work has identified spans in the sequence likely to contribute structurally; the C-terminal end of the transmembrane helix C, the N-terminal section of the cd loop, including the amphipathic cd-helix, the ef loop, including a coil from the end of the helix E, the -PEWY- turn, the small ef helix connecting the -PEWY- turn to transmembrane helix, and the N-terminal end of helix F. In the structure, these spans define two features: i) a quinol binding pocket, occupying a volume in which a difference electron density is seen in crystals containing myxothiazol or stigmatellin, and ii) a concave surface, which is a docking interface for the ISP. The transmembrane helices contributing to the site flare out to provide the volume, and to form the sides of the pocket, the -PEWY- loop acts as a spacer, and the amphipathic helices provide the bottom. They also, on their other faces, provide most of the ISP binding surface, with the ef-loop providing additional residues. From examination of the placing of these domains in the structure it is apparent that mutations in either can interfere with the catalytic mechanism. Differential effects on binding of the two classes of inhibitor acting at the site have been noted, with strains resistant to stigmatellin, but not myxothiazol or mucidin, and vice versa, and strains resistant to both inhibitors (1, 27, 28). Binding studies have shown that occupation is mutually exclusive (3, 10, 12, 13, 32-35), so that only one occupant (quinone, stigmatellin, myxothiazol, or UHDBT) can be in the site, and the crystallographic studies confirm these earlier results. Mutations have several different effects on the changes in EPR spectrum of the Rieske center at g x =1.800, which have been attributed to interaction with quinone at the Q o -site. However, for any particular mutation, the effects on signals attributed to Q ow and Q os have been the same. The Q o -site is shown in greater detail in Fig. 2, in which residues known to effect inhibitor binding or catalysis are shown as stick models. The locations of stigmatellin and myxothiazol are modeled on the basis of the electron density differences observed in separate crystals containing these inhibitors, compared to the native structure.

Figure 2. Residues at which mutation leads to modification of function. A. Inhibitor resistance sites. Note that in many cases, several mutations have been generated at a site, with differential effects on inhibitor binding, depending on the nature of the change. The following color-coding is therefore somewhat simplistic. Myxothiazol resistant sites are shown in yellow, stigmatellin resistance sites are shown in red. Residues at which modification leads to resistance to both inhibitors are shown in green. Stigmatellin and myxothiazol are shown as ball and stick models, colored cyan and white respectively, occupying the volumes in which they are found in separate crystals. The heme of cyt b L is shown as a magenta wireframe model. The iron sulfur protein is shown in the stigmatellin-induced configuration, as a blue backbone model. B. Residues at which mutation produces a modified g x =1.800 band. Yellow, normal occupancy but no rate; red, empty, but moderate rate; magenta, empty, and little or no rate; violet, partially occupied, slow rate, hyper sensitive to myxothiazol. Heme is orange. Inhibitors as ball and stick models; stigmatellin is green, myxothiazol is white. The iron sulfur protein (chain E in Berry's structure) is shown as a blue backbone model, with Leu-142 and Gly-143 colored gray and brown. Changes at these residues also modify the kinetics of the Q o -site, and the g x =1.800 signal. Kinetics in the high potential chain Myxothiazol binds at the Q o -site of cyt b, and displaces quinol, preventing the delivery of electrons to the 2Fe2S center of the ISP. This simplifies the electron transfer chain so that the reactions of the high potential components can be examined separately. In Rb. sphaeroides, oxidizing equivalents from the primary donor (P + /P) of photochemical reaction center (RC) are delivered to cytochrome c 1 by cytochrome c 2, with a t ½; of about 150 µs. The electron transfer from the Rieske center to cyt c 1 cannot be resolved, because it is not rate determining. However, kinetic arguments suggest that the rate constant must be in the range 10 5 s -1 (5, 7, 9). In the absence of myxothiazol, the bifurcated reaction occurs, resulting in cytochrome b reduction. A ~200 µs lag observed before onset of reduction of cyt b H is contributed by several processes: the delivery of oxidizing equivalents to cyt c 1 (~150 µs), the oxidation of the 2Fe2S center (<10 µs), and the movement of the ISP to its reaction interface with QH 2. The reactions of the ISP with its partners (QH 2 bound at the Q o -site of cyt b, and the heme of cyt c 1 ) (Fig. 1) require a movement of ~ 16 Å

between two reaction domains on these separate subunits. Kinetics of quinol oxidation In the oxidation of quinol, the kinetics of electron transfer to the high potential chain and the cytochrome b chain are closely matched. The system behaves as if the oxidation involved a concerted, two-electron reaction with simultaneous delivery of electrons to two separate chains. However, it is generally accepted that this behavior reflects a sequential reaction in which the intermediate semiquinone is consumed rapidly, and never attains a significant concentration. Q o H 2.(2Fe2S +.c 1 + ).(b L +.b H +.Q i ) = Q o.-.(2fe2s.c 1 ) +.(b L +.b H +.Q i ) + 2H P + (~600 µs) Q o.-.(2fe2s.c 1 ) +.(b L +.b H +.Q i ) = Q o.(2fe2s.c 1 ) +.(b L.b H +.Q i ) (< 60 µs) Overall: Q o H 2.(2Fe2S +.c 1 + ).(b L +.b H +.Q i ) = Q o.(2fe2s.c 1 ) +.(b L.b H +.Q i ) + 2H P + Because the reduction of cyt b L by Q o.-, and of cyt b H by cyt b L are not rate limiting (34, 36), the overall reaction can be assayed by measuring the rate of reduction of cyt b H in the presence of antimycin; quinol oxidation is the rate limiting step under conditions of saturation with substrate. Crofts and Wang (9) developed a model that accounted well for the kinetic features of quinol oxidation; although this provided an excellent fit to the kinetics over a wide range of conditions, one aspect of the mechanism of quinol oxidation was not explicitly address. It has been a long standing paradox that the bifurcated reaction at the Q o -site delivered electrons stoichiometrically to the two separate chains, despite the strong thermodynamic potential favoring delivery of both electrons to the high potential chain (see Brandt (37) for a recent discussion). Double-occupancy model Earlier work had shown an effect of the redox state of the quinone pool on the EPR spectrum of the 2Fe2S center of the complex (33, 38, 39). More recently, Ding et al. (34, 35, 40), using wildtype and mutant strains from Rb. capsulatus, have investigated the effects of extraction of ubiquinone, and concluded that different spectral changes can be detected at different local concentrations. They suggested that these reflect two different bound quinones at the Q o -site, called Q os and Q ow for strongly and weakly binding species, occupying separate domains, at both of which Q and QH 2 bind with equal affinity (i.e. with no change in E m with respect to the pool). They have extended these observations to mutant strains, and kinetic studies of turnover and inhibitor binding, and have discussed possible mechanisms and atomic details of ligation. Brandt (43) has recently expanded on the double occupancy model to suggest an explanation for the observation that electrons do not "leak" rapidly from the strongly reducing semiquinone at the Q o -site into the high potential chain. The extensive work on the g x signals of the 2Fe2S center has contributed a useful set of data on interactions of quinone and inhibitor species with the ISP, and on occupancy of the Q o -site. Nevertheless, there are some problems with the interpretation offered (28).

Effects of Q o -site occupancy on EPR spectra of the Iron Sulfur Protein. In summary, with the Q-pool oxidized and ISP reduced, a sharp band at g x =1.80 is seen. When the pool is reduced, or Q extracted, or myxothiazol bound at the Q o -site, the g x signal shifts from 1.80 to ~1.765, and becomes smaller (34, 35, 39, 40). When the pool is extracted so as to leave ~2 Q/RC (1 as Q A, plus ~2Q/bc 1 -complex in the preparations used) an intermediate signal at g x =1.783 is seen. Several possible explanations for these effects on the EPR spectrum can be offered: i. Ding et al. (35, 40) suggested that the g x =1.783 signal was due to the Q os species, and that this would likely be the one forming a strong interaction with the ISP. From the structure, this would translate to Q os occupying the distal end of the pocket (where stigmatellin binds). Q ow was suggested to be an exchangeable species, through which coupling to the quinone pool could occur. In the context of the structure, Q ow would be expected to bind at the proximal (myxothiazol) end, nearer heme b L. Since the g x =1.800 signal is ascribed to Q ow, the interaction with the 2Fe2S center indicated by the signal would have to be indirect, and mediated by Q os. ii. The g x =1.800 signal is due to ISP red interacting directly with Q at the Q o -site, and is lost when the site is either empty, occupied by inhibitor, or by QH 2. We believe this explanation requires the fewest ad hoc hypotheses. However, if this is the case, then under conditions in which the quinone pool is oxidized before flash-activation, the reduced ISP would not be in position for its reaction with cytochrome c 1. It would be necessary to postulate that the diffusion time between cyt b and cyt c 1 docking interfaces is rapid compared to the <10 µs oxidation time of 2Fe2S observed, and that the binding at the cyt b interface under these conditions (Q oxidized, ISP reduced) must not be so great as to prevent the ISP leaving within this time. We discuss these parameters in greater detail elsewhere. (31). In deciding between these explanations, the following points should also be considered: i. None of the structures (22, 23) show a density in the uninhibited site expected for a tightly bound quinone species. In the Berry structures, a weak density in the unoccupied site is observed, which may represent a quinone species, either weakly bound with low occupancy in a fixed position, or mobile in the site. In the Xia et al. structure, the authors reported that no loss of density from the pocket was observed on binding inhibitors at the Q o -site, although a loss of density was observed at the Q i -site on binding antimycin, and this was attributed to displacement of quinone (23). Since their crystals contained substoichiometric levels of quinone (~0.6 Q/complex), these results would suggest that the Q i -site binds the quinone more tightly than the Q o -site. ii. In the structures from both groups, the electron densities of inhibitors in the stigmatellin and myxothiazol crystals occupy overlapping volumes, suggesting, in line with competition studies, that these two inhibitors are mutually exclusive occupants of the site (see Fig. 2). Although at the current structural resolution, we cannot exclude the possibility that two ubiquinones could occupy the same site, it seems probable that only one quinone can occupy this volume.

iii. The intermediate line-shape at g x =1.783, seen when ~2 Q / bc 1 -complex are present, is attributed to Q os, a species that binds 30-fold more tightly than Q ow (34, 35). Paradoxically, the other properties of these two species are similar. This paradox could be resolved if the line shape change reflected only one species. In attributing the 1.783 signal, Dutton and colleagues assumed a tight binding species. A difficulty with this assumption comes from the heterogeneity in distribution of redox components likely in a population of chromatophores (cf. 42). Extraction of chromatophores to the point giving rise to the Q os signal would leave the distribution of quinone random, so that some chromatophores would have an excess of quinone over the 2 per bc 1 -complex, and we would expect to see a sharp peak at 1.800 due to Q ow. In another fraction, the quinone would be below this ratio, leaving some bc 1 -complex without a quinone, with a shallow peak at 1.765. At the very least, we would expect in the overall population a peak representing the convolution of these two spectra, at a position intermediate between them, as previously seen in pentane extracted mitochondrial complex III by de Vries et al. (39). iv. The environment of the 2Fe2S center, and the protein interactions of the Rieske subunit, will change substantially as the ISP moves. In addition, the interaction between the spins of the reduced 2Fe2S center, and the oxidized cyt b L (52) might also be expected to change during catalysis, since the distance between them would change by ~8 Å. Some perturbation of the spectrum of the 2Fe2S center might be attributed to these changes. We suggest a simpler interpretation of the g x =1.800 signal and the changes seen on reduction, extraction, or in mutant strains. a. The g x =1.800 signal is associated with a complex formed between quinone bound in the Q o -site at the distal end, and the reduced ISP docked firmly at the interface with cytochrome b. b. The signal is lost whenever this complex cannot form: - on reduction of Q, on binding of inhibitors, on extraction of quinone, in strains where mutation prevents binding at the distal end of the pocket, and in strains where mutation prevents docking of the ISP. c. Only one quinone species occupies the site. The spectroscopic effects (g x =1.783) attributed to Q os arise from the statistical distribution of quinone in the heterogeneous chromatophore population, with additional contributions from changes in environment of the ISP due to changes in its interactions accompanying extraction of the last occupant, to leave a vacant site. As an alternative to c), a modified double-occupancy model might be suggested in which Q ow occupies the distal site, and Q os the proximal site, and the latter serves as an electron shuttle to heme b L on formation of the semiquinone on the former. The paradox of the bifurcated reaction In the presence of antimycin, the Q o -site is free to turn over, and under steady state conditions in which quinol is available in the pool, the b-cytochrome chain becomes reduced, and the ISP is oxidized by the high potential acceptor. Under these circumstances, de Vries was able to detect a small amount of semiquinone at the Q o -site. Since antimycin is an effective inhibitor, and net electron transfer is blocked under these conditions, the semiquinone must not pass its second

electron to the ISP. How is this thermodynamically favored reaction prevented? Several factors might be involved: i. a set of forces in the binding domain which disfavor the binding of the oxidized ISP, or ii. iii. displacement of the semiquinone by ligand exchange to a position deeper in the pocket, and closer to its reaction partner, the cyt b L heme, or a conformational change in which movement of one or more residues "insulates" the quinone binding pocket from the ISP domain. These are not mutually exclusive; indeed some contribution from each is likely to occur, since the liganding interaction between Q o -site occupant and the ISP will contribute to the force favoring occupancy, and any movement of the occupant of the site will likely involve some rearrangement of side chains. We favor the second of these possibilities as the main factor. The possibility of such a movement is supported by the deeper position in the pocket of myxothiazol compared to stigmatellin (Fig. 2). The recent evidence for a substantial movement of Q B in its binding pocket when it is reduced to semiquinone in the bacterial reaction center (41), also shows the possibility of such movement. Stowell et al. suggest that the activation barrier which gives the high temperature dependence of electron transfer from Q A - Q B to Q A Q B -, might reflect this movement, and such considerations could obviously be applied to the reaction at the Q o -site. In support of the third possibilitiy, the position of the head of stigmatellin (as currently modeled) overlaps the volume occupied by the ring of a tyrosine side chain (Tyr-279) in the uninhibited complex, and preliminary analysis suggests that the sidechain rotates from this position on binding of inhibitor (Berry, E.A., unpublished). In the absence of inhibitor, the tyrosine sits at the opening between the Q-binding pocket and the ISP interface. It seems likely that any occupant of the pocket that interacts with the ISP can only do so if the tyrosine is displaced. We suggest that the tyrosine might act as a trap-door controlling access to the occupant of the binding pocket. Stigmatellin also occupies some common volume with the position of the ring of Pro-271 in the uninhibited structure, which also appears to be involved in a small conformational change on occupation of the distal end of the pocket. The distribution of residues at which mutation results in modified function: distinct domains and modes of action The extensive set of data provided by Ding et al. (34, 35, 40) on occupancy of the site in different mutant strains, as reflected in the g x =1.800 signal of the 2Fe2S center, and our own work in collaboration with these authors (28), provide important clues about the mechanism. At the present resolution of the structure, we can distinguish several different classes of mutational change, which are color-coded in Fig. 2A (inhibitor resistance sites) and B (residues where change leads to a modified g x =1.800 signal). i. Residue changes at the interface with the ISP Changes at Lys-288, Leu-282, Thr-145, Gly-143, Ile-269, which can be seen to project into the ISP interfacial surface in the Fig. 2B, eliminate the g x =1.800 signal, which we have attributed to interaction between quinone at the Q o -site and the reduced ISP. In

some cases this is associated with a loss of activity, but some of these strains show substantial turnover. It seems possible that the mutations that eliminate or reduce the g x signal interfere with the ISP binding, and prevent formation of the complex between Q and ISP red that gives rise to this signal. The steric effects that give rise to loss of the EPR band might also be expected to interfere with the access of the ISP ox to QH 2 at the site. The variable affects on turnover number would then be explained by the decrease in the residence time of the ISP at the docking interface, and therefore the probability of formation of the reaction complex. ii. Residue changes at the proximal end of the Q o -binding pocket Residue changes (yellow in Fig. 2B above), which lead to loss of activity without loss of the g x =1.800 signal, cluster at the proximal end of the pocket, closer to the heme of cyt b L, where myxothiazol binds. Many of the myxothiazol resistance strains also have residue changes at this end of the pocket (yellow and green in Fig. 2A). It seems reasonable to conclude that these mutations do not interfere with the binding of either Q at the distal end, or the ISP. However, they do prevent oxidation of QH 2. The inhibition of oxidation in these mutants must reflect some effect relating to the occupancy of the proximal end of the pocket. This could either be a second quinone species involved in electron transfer to the b L heme (in a modified double-occupancy model), or a need for movement of the semiquinone to the proximal position to bring it closer to cyt b L to allow rapid electron transfer. iii. Residue changes at the distal end of the Q o -binding pocket Consistent with the model developed above, changes to residues impinging on the stigmatellin binding domain lead to loss of the g x =1.800 signal (red and magenta in Fig. 2B), and variable effects on electron transfer rate and inhibitor binding. In general, as might be expected, many of the mutational changes giving resistance to stigmatellin are found in this set (red and green residues in Fig. 2A). Conclusions We suggest as an alternative to the double-occupancy model, the following mechanism for turnover of the Q o -site of the complex: a. Oxidizing equivalents are transferred to the site through cytochrome c 1 and the ISP. The mechanism involves a movement of the ISP between reaction domains on cytochrome c 1 and cytochrome b (Fig.1). b. The reaction proceeds from a complex between the oxidized ISP and quinol. The reaction complex involves QH 2 bound at the distal end of the pocket, and the oxidized ISP docked tightly at the interface on cytochrome b. Binding of QH 2 and formation of the complex requires a displacement of Tyr-279, to allow access of the ISP to the quinol. This binding likely also leads to release of 1 H + through an interaction at the site that changes the ef-

fective pk of the QH 2 /QH -.H + dissociation reaction. c. Electron transfer from QH 2 to the ISP leads to formation of semiquinone. d. The semiquinone moves in the pocket to the proximal end, near heme b L. e. The Tyr-279 trap-door flips back to its "closed" position, insulating the semiquinone from further reaction with the ISP. f. The semiquinone passes its electron to the cyt b L heme, and the quinone exits the site. g. At some point between c) and f), the second proton is released. This might be required for formation of the semiquinone, or its movement in the pocket, or the transfer of the second electron. We suggest that the ph dependence of the activation barrier (37) reflects this second deprotonation, and that the activation barrier is in one of these steps. h. Meanwhile, formation of the semiquinone liberates the reduced ISP so that it can deliver an electron to cytochrome c 1 by the tethered diffusion mechanism we have previously suggested. Acknowledgments We are grateful to Professor Thomas Link for kindly providing us with the coordinates of the ISP soluble fragment before these were publicly available. We acknowledge with gratitude the support for this research provided by NIH grants GM 35438 (to ARC) and DK 44842 (to EAB), and by the Office of Health and Environmental Research, US Department of Energy, under contract DE-AC03-76SF00098 (EAB). The work was partially done at SSRL which is operated by the Department of Energy, Division of Chemical/Material Sciences. The SSRL Biotechnology Program is supported by the National Institutes of Health Biomedical Resource Technology Program, Division of Research Resources. Reference 1. Gennis, R.B., Barquera, B., Hacker, B., van Doren, S.R., Arnaud, S., Crofts, A.R., Davidson, E., Gray, K.A. and Daldal, F. (1993) J. Bioenerg. Biomembr. 25, 195-210. 2. Brandt, U. and Trumpower, B.L. (1994) CRC Crit. Rev. Biochem. 29, 165-197. 3. Schägger, H., Brandt, U., Gencic, S. and von Jagow, G. (1995) Methods Enzymol. 260, 82-96. 4. Mitchell, P. (1976) J. Theor. Biol. 62, 327-367 5. Crofts, A. R., Meinhardt, S. W., Jones, K. R. and Snozzi, M. (1983) Biochim. Biophys. Acta, 723, 202-218 6. Glaser, E.G. and Crofts, A.R. (1984). Biochim. Biophys. Acta, 766, 322-333. 7. Crofts, A. R. (1985). In: The Enzymes of Biological Membranes, (Martonosi, A.N., ed.), Vol. 4, pp. 347-382, Plenum Publ. Corp., New York. 8. Robertson, D.E. and Dutton, P.L. (1988) Biochim. Biophys. Acta 935, 273-291. 9. Crofts, A.R. and Wang, Z. (1989) Photosynth. Res. 22, 69-87

10. Bowyer, J. R., Dutton, P. L., Prince, R. C., & Crofts, A. R. (1980) Biochim. Biophys. Acta 592, 445-460. 11. Meinhardt, S.W. and Crofts, A.R. (1982). FEBS Lett., 149, 217-222. 12. Link, T.A., Haase, U., Brandt, U. and von Jagow, G. (1993) J. Bioenerg. Biomemb. 25, 221-232 13. von Jagow, G. and Link, T.A. (1986) Methods Enzymol. 126, 253-271 14. Crofts, A. R. and Wraight, C. A. (1983). Biochim. Biophys. Acta, 726, 149-186. 15. Iwata, S., Saynovits, M., Link, T.A. and Michel, H. (1996) Structure 1996, 4: 567-579. 16. Xia, D., Yu, C.-A., Deisenhofer, J., Xia, J.-Z. and Yu, L. (1996) Abstracts. Biophys. Soc. Meeting, Baltimore, Feb. 1996. 17. Xia, D., Kim, H., Deisenhofer, J., Yu, C.-A., Xia, J.-Z. and Yu, L. (1996) Abstracts, #SO355. XVII Meeting, Intntl. Union Crystallography, Seattle. 18. Kim, H., Xia, D., Deisenhofer, J., Yu, C.-A., Kachurin, A. and Yu, L. (1996) Abstracts, #SO356. XVII Meeting, Intntl. Union Crystallography, Seattle. 19. Yu, C.-A., Xia, J.-Z., Kachurin, A.M., Yu, L., Xia, D., Kim, H. and Deisenhofer, J. (1996) Biochim. Biophys. Acta 1275, 47-53. 20. Berry, E.A., Zhang, Z., Huang, L.-S., Chi, Y.-I., and Kim, S.-H. (1997) Abstracts, Biophy. Soc. Ann. Meeting, New Orleans, March 1997, Tu-PM-G4 21. Berry, E.A., Shulmeister, V.M., Huang, L.S. and Kim, S. H. (1995) Acta Crystallographica 51, 235-239. 22. Zhang, Z., Huang, L., Chi, Y.-I., Kim, K.K., Crofts, A.R., Berry, E.A., and Kim, S.-H. (1997) Manuscript in preparation. 23. Xia, D., Yu, C.-A., Kim, H., Xia, J.-Z., Kachurin, A.M., Zhang, L., Yu, L., Deisenhofer, J. (1997) Science, 277, 60-66 24. Yun, C.-H., Beci, R., Crofts, A.R., Kaplan, S. and Gennis, R.B. (1990) Europ. J. Biochem. 194, 399-411 25. Yun, C.-H., Crofts, A.R. and Gennis, R.B. (1991) Biochemistry, 30, 6747-6754 26. Konishi, K., Van Doren, S.R., Kramer, D.M., Crofts, A.R., and Gennis, R.B. (1991) J. Biol. Chem. 266, 14270-14276 27. Brasseur, G., Sami Saribas, A. and Daldal, F. (1996) Biochim. Biophys. Acta 1275, 61-69. 28. Crofts, A.R., Barquera, B., Bechmann, G., Guergova, M, Salcedo-Hernandez, R., Hacker, B., Hong, S. and Gennis, R.B. (1995) In Photosynthesis: from light to biosphere. (Mathis, P., ed.), Vol. II, pp. 493-500. Kluwer Academic Publ., Dordrecht. 29. Crofts, A.R., Hacker, B., Barquera, B., Yun, C.-H. and Gennis, R. (1992) Biochim. Biophys. Acta 1101, 162-165 30. Degli Esposti, M., De Vries, S., Crimi, M., Ghelli, A., Patarnello, T., and Meyer, A. (1993) Biochim. Biophys. Acta 1143, 24-271 31. Berry, E.A., Zhang, Z., Huang, L.-S., Kuras, R., Guergova-Kuras, M. and Crofts, A.R. (1997) Abstract, Meeting on "Reaction centers of photosynthetic purple bacteria: structure, spectroscopy, dynamics", Cadarache, June 1997 32. Meinhardt, S.W. and Crofts, A.R. (1982). FEBS Lett. 149, 223-227. 33. Matsuura, K., Bowyer, J.R., Ohnishi, T., & Dutton, P.L. (1983) J. Biol. Chem. 258, 1571-1579. 34. Ding, H., Robertson, D.E., Daldal, F. and Dutton, P.L. (1992) Biochemistry 31, 3144-3158.

35. Ding, H., Moser, C.C., Robertson, D.E., Tokito, M.K., Daldal, F. and Dutton, P.L. (1995) Biochemistry 34, 15979-15996. 36. Glaser, E.G., Meinhardt, S.W. and Crofts, A.R. (1984) FEBS Lett. 178, 336-342 37. Brandt, U. (1996) Biochim. Biophys. Acta 1275, 41-46. 38. Siedow, J.N., Power, S., De La Rosa, F.F. and Palmer, G. (1978) J. Biol. Chem. 253, 2392-2399 39. De Vries, S., Albracht, S.P.J., Berden, J. and Slater, E.C. (1982) Biochim. Biophys. Acta 681, 41-53 40. Ding, H., Daldal, F. and Dutton, P.L. (1995) Biochemistry 34, 15997-16003 41. Stowell, M. H. B., McPhillips, T. M., Rees, D. C., Soltis, S. M., Abresch, E. and Feher, G. (1997) Science, 276, 812-815 42. Crofts, A.R., Guergova-Kuras, M., Hong, S. and Bechmann, G. (1997) Photosynth. Research, in press.