首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The vacuolar H+-ATPase (V-ATPase) is an ATP-driven proton pump essential to the function of eukaryotic cells. Its cytoplasmic V1 domain is an ATPase, normally coupled to membrane-bound proton pump Vo via a rotary mechanism. How these asymmetric motors are coupled remains poorly understood. Low energy status can trigger release of V1 from the membrane and curtail ATP hydrolysis. To investigate the molecular basis for these processes, we have carried out cryo-electron microscopy three-dimensional reconstruction of deactivated V1 from Manduca sexta. In the resulting model, three peripheral stalks that are parts of the mechanical stator of the V-ATPase are clearly resolved as unsupported filaments in the same conformations as in the holoenzyme. They are likely therefore to have inherent stiffness consistent with a role as flexible rods in buffering elastic power transmission between the domains of the V-ATPase. Inactivated V1 adopted a homogeneous resting state with one open active site adjacent to the stator filament normally linked to the H subunit. Although present at 1:1 stoichiometry with V1, both recombinant subunit C reconstituted with V1 and its endogenous subunit H were poorly resolved in three-dimensional reconstructions, suggesting structural heterogeneity in the region at the base of V1 that could indicate positional variability. If the position of H can vary, existing mechanistic models of deactivation in which it binds to and locks the axle of the V-ATPase rotary motor would need to be re-evaluated.  相似文献   

2.
Bovine V-ATPase from brain clathrin-coated vesicles was investigated by cryo-electron microscopy and single particle analysis. Our studies revealed great flexibility of the central linker region connecting V1 and V0. As a consequence, the two sub-complexes were processed separately and the resulting volumes were merged computationally. We present the first three-dimensional (3D) map of a V-ATPase obtained from cryo-electron micrographs. The overall resolution was estimated 34 Å by Fourier shell correlation (0.5 cutoff). Our 3D reconstruction shows a large peripheral stalk and a smaller, isolated peripheral density, suggesting a second, less well-resolved peripheral connection. The 3D map reveals new features of the large peripheral stator and of the collar-like density attached to the membrane domain. Our analyses of the membrane domain indicate the presence of six proteolipid subunits. In addition, we could localize the V0 subunit a flanking the large peripheral stalk.  相似文献   

3.
The Na+-pumping V-ATPase complex of the thermophilic bacterium Caloramator fervidus was purified and dissociated under controlled conditions. The structure of purified V1-ATPase subcomplexes differing in subunit composition was analyzed by electron microscopy and single particle analysis of 50 000 projections. Difference mapping of subcomplex projections revealed the presence and position of two subunits in the central stalk. A density with an elongated shape similar to the γ subunit of F-ATPases is partly located within V1 and corresponds, most likely, to subunit E. Subunit E is connected to the membrane-bound part V0 via subunit C, a spherical density that is connected to the center of V0. The presence of subunit C makes the central stalk substantially longer in comparison to the F-ATPases, in which the γ subunit connects directly to F0.  相似文献   

4.

Background

Vacuolar (H+)-ATPase (V-ATPase; V1Vo-ATPase) is a large multisubunit enzyme complex found in the endomembrane system of all eukaryotic cells where its proton pumping action serves to acidify subcellular organelles. In the plasma membrane of certain specialized tissues, V-ATPase functions to pump protons from the cytoplasm into the extracellular space. The activity of the V-ATPase is regulated by a reversible dissociation mechanism that involves breaking and re-forming of protein-protein interactions in the V1-ATPase - Vo-proton channel interface. The mechanism responsible for regulated V-ATPase dissociation is poorly understood, largely due to a lack of detailed knowledge of the molecular interactions that are responsible for the structural and functional link between the soluble ATPase and membrane bound proton channel domains.

Methodology/Principal Findings

To gain insight into where some of the stator subunits of the V-ATPase associate with each other, we have developed peptide arrays from the primary sequences of V-ATPase subunits. By probing the peptide arrays with individually expressed V-ATPase subunits, we have identified several key interactions involving stator subunits E, G, C, H and the N-terminal domain of the membrane bound a subunit.

Conclusions

The subunit-peptide interactions identified from the peptide arrays complement low resolution structural models of the eukaryotic vacuolar ATPase obtained from transmission electron microscopy. The subunit-subunit interaction data are discussed in context of our current model of reversible enzyme dissociation.  相似文献   

5.
Li Z  Zhang X 《Planta》2004,219(6):948-954
The vacuolar H+-ATPase from mung bean (Vigna radiata L. cv. Wilczek) was purified to homogeneity. The purified complex contained all the reported subunits from mung bean, but also included a 40-kDa subunit, corresponding to the membrane-associated subunit d, which has not previously been observed. The structure of the V-ATPase from mung bean was studied by electron microscopy of negatively stained samples. An analysis of over 6,000 single-particle images obtained by electron microscopy of the purified complex revealed that the complex, similar to other V-ATPases, is organized into two major domains V1 and Vo with overall dimensions of 25 nm×13.7 nm and a stalk region connecting the V1 and Vo domains. Several individual areas of protein density were observed in the stalk region, indicating its complexity. The projections clearly showed that the complex contained one central stalk and at least two peripheral stalks. Subcomplexes containing subunits A, B and E, dissociated from the tonoplast membrane by KI, were purified. The structure of the subcomplex was also studied by electron microscopy followed by single-molecule analysis of 13,000 projections. Our preliminary results reveal an area of high protein density at the bottom of the subcomplex immediately below the cavity formed by the A and B subunits, indicating the position of subunit E.Abbreviations MSA Multivariate statistical analysis - 2D, 3D Two-, three-dimensional - V-ATPase Vacuolar H+-ATPase  相似文献   

6.
The yeast vacuolar H+-ATPase (V-ATPase) is a multisubunit complex responsible for organelle acidification. The enzyme is structurally organized into two major domains: a peripheral domain (V1), containing the ATP binding sites, and an integral membrane domain (V0), forming the proton pore. Dissociation of the V1 and V0 domains inhibits ATP-driven proton pumping, and extracellular glucose concentrations regulate V-ATPase activity in vivo by regulating the extent of association between the V1 and V0 domains. To examine the mechanism of this response, we quantitated the extent of V-ATPase assembly in a variety of mutants with known effects on other glucose-responsive processes. Glucose effects on V-ATPase assembly did not involve the Ras-cyclic AMP pathway, Snf1p, protein kinase C, or the general stress response protein Rts1p. Accumulation of glucose 6-phosphate was insufficient to maintain or induce assembly of the V-ATPase, suggesting that further glucose metabolism is required. A transient decrease in ATP concentration with glucose deprivation occurs quickly enough to help trigger disassembly of the V-ATPase, but increases in cellular ATP concentrations with glucose readdition cannot account for reassembly. Disassembly was inhibited in two mutant enzymes lacking ATPase and proton pumping activities or in the presence of the specific V-ATPase inhibitor, concanamycin A. We propose that glucose effects on V-ATPase assembly occur by a novel mechanism that requires glucose metabolism beyond formation of glucose 6-phosphate and generates a signal that can be sensed efficiently only by a catalytically competent V-ATPase.  相似文献   

7.
Vacuolar ATPases (V-ATPases) are molecular machines responsible for creating electrochemical gradients and preserving pH-dependent cellular compartments by way of proton translocation across the membrane. V-ATPases employ a dynamic rotary mechanism that is driven by ATP hydrolysis and the central rotor stalk. Regulation of this rotational catalysis is the result of a reversible V1Vo-domain dissociation that is required to preserve ATP during instances of cellular starvation. Recently the method by which the free V1-ATPase abrogates the hydrolytic breakdown of ATP upon dissociating from the membrane has become increasingly clear. In this instance the central stalk subunit F adopts an extended conformation to engage in a bridging interaction tethering the rotor and stator components together. However, the architecture by which this mechanism is stabilized has remained ambiguous despite previous work. In an effort to elucidate the method by which the rotational catalysis is maintained, the architecture of the peripheral stalks and their respective binding interactions was investigated using cryo-electron microscopy. In addition to confirming the bridging interaction exuded by subunit F for the first time in a eukaryotic V-ATPase, subunits C and H are seen interacting with one another in a tight interaction that provides a base for the three EG peripheral stalks. The formation of a CE3G3H sub-assembly appears to be unique to the dissociated V-ATPase and highlights the stator architecture in addition to revealing a possible intermediate in the assembly mechanism of the free V1-ATPase.  相似文献   

8.
The RAVE complex (regulator of the H+-ATPase of vacuolar and endosomal membranes) is required for biosynthetic assembly and glucose-stimulated reassembly of the yeast vacuolar H+-ATPase (V-ATPase). Yeast RAVE contains three subunits: Rav1, Rav2, and Skp1. Rav1 is the largest subunit, and it binds Rav2 and Skp1 of RAVE; the E, G, and C subunits of the V-ATPase peripheral V1 sector; and Vph1 of the membrane Vo sector. We identified Rav1 regions required for interaction with its binding partners through deletion analysis, co-immunoprecipitation, two-hybrid assay, and pulldown assays with expressed proteins. We find that Skp1 binding requires sequences near the C terminus of Rav1, V1 subunits E and C bind to a conserved region in the C-terminal half of Rav1, and the cytosolic domain of Vph1 binds near the junction of the Rav1 N- and C-terminal halves. In contrast, Rav2 binds to the N-terminal domain of Rav1, which can be modeled as a double β-propeller. Only the V1 C subunit binds to both Rav1 and Rav2. Using GFP-tagged RAVE subunits in vivo, we demonstrate glucose-dependent association of RAVE with the vacuolar membrane, consistent with its role in glucose-dependent V-ATPase assembly. It is known that V1 subunit C localizes to the V1-Vo interface in assembled V-ATPase complexes and is important in regulated disassembly of V-ATPases. We propose that RAVE cycles between cytosol and vacuolar membrane in a glucose-dependent manner, positioning V1 and V0 subcomplexes and orienting the V1 C subunit to promote assembly.  相似文献   

9.
The proton (H+) pumping vacuolar-type ATPase (V-ATPase) is a rotary enzyme that plays a pivotal role in forming intracellular acidic compartments in eukaryotic cells. In Saccharomyces cerevisiae, the membrane extrinsic catalytic V1 and the transmembrane proton-pumping Vo complexes have been shown to reversibly dissociate upon removal of glucose from the medium. However, the basis of this disassembly is largely unknown. In the earlier study, we have found that the amino-terminal α-helical domain between Lys-33 and Lys-83 of yeast E subunit (Vma4p) in the peripheral stalk of the V1 complex has a role in glucose-dependent VoV1 assembly. Results of alanine-scanning mutagenesis within the domain revealed that the Vma4p Glu-44 is a key residue in VoV1 disassembly. Biochemical analysis on Vma4p Glu-44 to Ala, Asn, Asp, and Gln substitutions indicated that Glu-44 has a role in V-ATPase catalysis. These results suggest that Glu-44 is one of the key functional residues for subunit interaction in the V-ATPase stalk complex that allows both efficient rotation catalysis and assembly.  相似文献   

10.
The vacuolar (H+)-ATPase: subunit arrangement and in vivo regulation   总被引:1,自引:0,他引:1  
The V-ATPases are responsible for acidification of intracellular compartments and proton transport across the plasma membrane. They play an important role in both normal processes, such as membrane traffic, protein degradation, urinary acidification, and bone resorption, as well as various disease processes, such as viral infection, toxin killing, osteoporosis, and tumor metastasis. V-ATPases contain a peripheral domain (V1) that carries out ATP hydrolysis and an integral domain (V0) responsible for proton transport. V-ATPases operate by a rotary mechanism involving both a central rotary stalk and a peripheral stalk that serves as a stator. Cysteine-mediated cross-linking has been used to localize subunits within the V-ATPase complex and to investigate the helical interactions between subunits within the integral V0 domain. An essential property of the V-ATPases is the ability to regulate their activity in vivo. An important mechanism of regulating V-ATPase activity is reversible dissociation of the complex into its component V1 and V0 domains. The dependence of reversible dissociation on subunit isoforms and cellular environment has been investigated. Qi and Wang contributed equally to this work.  相似文献   

11.
The prokaryotic V-type ATPase/synthases (prokaryotic V-ATPases) have simpler subunit compositions than eukaryotic V-ATPases, and thus are useful subjects for studying chemical, physical and structural properties of V-ATPase. In this review, we focus on the results of recent studies on the structure/function relationships in the V-ATPase from the eubacterium Thermus thermophilus. First, we describe single-molecule analyses of T. thermophilus V-ATPase. Using the single-molecule technique, it was established that the V-ATPase is a rotary motor. Second, we discuss arrangement of subunits in V-ATPase. Third, the crystal structure of the C-subunit (homolog of eukaryotic d-subunit) is described. This funnel-shape subunit appears to cap the proteolipid ring in the V0 domain in order to accommodate the V1 central stalk. This structure seems essential for the regulatory reversible association/dissociation of the V1 and the V0 domains. Last, we discuss classification of the V-ATPase family. We propose that the term prokaryotic V-ATPases should be used rather than the term archaeal-type ATPase (A-ATPase).  相似文献   

12.
Enterococcus hirae vacuolar ATPase (V-ATPase) is composed of a soluble catalytic domain (V1; NtpA3-B3-D-G) and an integral membrane domain (V0; NtpI-K10) connected by a central and peripheral stalk(s) (NtpC and NtpE-F). Here we examined the nucleotide binding of NtpA monomer, NtpB monomer or NtpD-G heterodimer purified by using Escherichia coli expression system in vivo or in vitro, and the reconstitution of the V1 portion with these polypeptides. The affinity of nucleotide binding to NtpA was 6.6 μM for ADP or 3.1 μM for ATP, while NtpB or NtpD-G did not show any binding. The NtpA and NtpB monomers assembled into NtpA3-B3 heterohexamer in nucleotide binding-dependent manner. NtpD-G bound NtpA3-B3 forming V1 (NtpA3-B3-D-G) complex independent of nucleotides. The V1 formation from individual NtpA and NtpB monomers with NtpD-G heterodimer was absolutely dependent on nucleotides. The ATPase activity of reconstituted V1 complex was as high as that of native V1-ATPase purified from the V0V1 complex by EDTA treatment of cell membrane. This in vitro reconstitution system of E. hirae V1 complex will be valuable for characterizing the subunit-subunit interactions and assembly mechanism of the V1-ATPase complex.  相似文献   

13.
Projection maps of a V1-Vma5p hybrid complex, composed of subunit C (Vma5p) of Saccharomyces cerevisiae V-ATPase and the C-depleted V1 from Manduca sexta, were determined from single particle electron microscopy. V1-Vma5p consists of a headpiece and an elongated wedgelike stalk with a 2.1×3.0 nm protuberance and a 9.5×7.5 globular domain, interpreted to include Vma5p. The interaction face of Vma5p in V1 was explored by chemical modification experiments.  相似文献   

14.
Disassembly of the yeast V-ATPase into cytosolic V1 and membrane V0 sectors inactivates MgATPase activity of the V1-ATPase. This inactivation requires the V1 H subunit (Parra, K. J., Keenan, K. L., and Kane, P. M. (2000) J. Biol. Chem. 275, 21761–21767), but its mechanism is not fully understood. The H subunit has two domains. Interactions of each domain with V1 and V0 subunits were identified by two-hybrid assay. The B subunit of the V1 catalytic headgroup interacted with the H subunit N-terminal domain (H-NT), and the C-terminal domain (H-CT) interacted with V1 subunits B, E (peripheral stalk), and D (central stalk), and the cytosolic N-terminal domain of V0 subunit Vph1p. V1-ATPase complexes from yeast expressing H-NT are partially inhibited, exhibiting 26% the MgATPase activity of complexes with no H subunit. The H-CT domain does not copurify with V1 when expressed in yeast, but the bacterially expressed and purified H-CT domain inhibits MgATPase activity in V1 lacking H almost as well as the full-length H subunit. Binding of full-length H subunit to V1 was more stable than binding of either H-NT or H-CT, suggesting that both domains contribute to binding and inhibition. Intact H and H-CT can bind to the expressed N-terminal domain of Vph1p, but this fragment of Vph1p does not bind to V1 complexes containing subunit H. We propose that upon disassembly, the H subunit undergoes a conformational change that inhibits V1-ATPase activity and precludes V0 interactions.V-ATPases are ubiquitous proton pumps responsible for compartment acidification in all eukaryotic cells (1, 2). These pumps couple hydrolysis of cytosolic ATP to proton transport into the lysosome/vacuole, endosomes, Golgi apparatus, clathrin-coated vesicles, and synaptic vesicles. Through their role in organelle acidification, V-ATPases are linked to cellular functions as diverse as protein sorting and targeting, zymogen activation, cytosolic pH homeostasis, and resistance to multiple types of stress (3). They are also recruited to the plasma membrane of certain cells, where they catalyze proton export (4, 5).V-ATPases are evolutionarily related to ATP synthases of bacteria and mitochondria and consist of two multisubunit complexes, V1 and V0, which contain the sites for ATP hydrolysis and proton transport, respectively. Like the ATP synthase (F-ATPase), V-ATPases utilize a rotational catalytic mechanism. ATP binding and hydrolysis in the three catalytic subunits of the V1 sector generate sequential conformational changes that drive rotation of a central stalk (68). The central stalk subunits are connected to a ring of proteolipid subunits in the V0 sector that bind protons to be transported. The actual transport is believed to occur at the interface of the proteolipids and V0 subunit a. Rotational catalysis will be productive in proton transport only if V0 subunit a is held stationary, whereas the proteolipid ring rotates (8). This “stator function” resides in a single peripheral stalk in F-ATPases (9, 10), but is distributed among up to three peripheral stalks in V-ATPases (1113). The peripheral stator stalks link V0 subunit a to the catalytic headgroup and ensures that there is rotation of the central stalk complex relative to the V0 a subunit and catalytic headgroup.Eukaryotic V-ATPases are highly conserved in both their overall structure and the sequences of individual subunits. Although homologs of most subunits of eukaryotic V-ATPases are present in archaebacterial V-ATPases (also known as A-ATPases), the C and H subunits are unique to eukaryotes. Both subunits have been localized at the interface of the V1 and V0 sectors, suggesting that they are positioned to play a critical role in structural and functional interaction between the two sectors (1416). The yeast C and H subunits are the only eukaryotic V-ATPase subunits for which x-ray crystal structures are available (17, 18). The structure of the C subunit revealed an elongated “dumbbell-shaped” molecule, with foot, head, and neck domains (18). The structure of the H subunit indicated two domains. The N-terminal 348 amino acids fold into a series of HEAT repeats and are connected by a 4-amino acid linker to a C-terminal domain containing amino acids 352–478 (17). These two domains have partially separable functions in the context of the assembled V-ATPase (19). Complexes containing only the N-terminal domain of the H subunit (H-NT)2 supported some ATP hydrolysis but little or no proton pumping in isolated vacuolar vesicles (19, 20). The C-terminal domain (H-CT) assembled with the rest of the V-ATPase in the absence of intact subunit H, but supported neither ATPase nor proton pumping activity (19). However, co-expression of the H-NT and H-CT domains results in assembly of both sectors with the V-ATPase and allows increased ATP-driven proton pumping in isolated vacuolar vesicles. These results suggest that the H-NT and H-CT domains play distinct and complementary roles even when the two domains are not covalently attached.In addition to their role as dedicated proton pumps, eukaryotic V-ATPases are also distinguished from F-ATPases and archaeal V-ATPases in their regulation. Eukaryotic V-ATPases are regulated in part by reversible disassembly of the V1 complex from the V0 complex (1, 21, 22). In yeast, disassembly of previously assembled complexes occurs in response to glucose deprivation, and reassembly is rapidly induced by glucose readdition to glucose-deprived cells. Disassembly down-regulates pump activity, and both the disassembled sectors are inactivated. Inhibition of ATP hydrolysis in free V1 sectors is particularly critical, because release of an active ATPase into the cytosol could deplete cytosolic ATP stores. This inhibition is dependent in part on the H subunit. V1 complexes isolated from vma13Δ mutants, which lack the H subunit gene (V1(-H) complexes) have MgATPase activity. Consistent with a physiological role for H subunit inhibition of V1, heterozygous diploids containing elevated levels of free V1 complexes without subunit H have severe growth defects (23). V1 complexes containing subunit H have no MgATPase activity, but retain some CaATPase activity, suggesting a role for nucleotides in inhibition (24, 25). Consistent with such a role, both the CaATPase activity of native V1 and the MgATPase activity of V1(-H) complexes are lost within a few minutes of nucleotide addition (24).A number of points of interaction between the H subunit and the V1 and V0 complexes have been identified through two-hybrid assays, binding of expressed proteins, and cross-linking experiments. These experiments have indicated that the H subunit binds to V1 subunits E and G of the V-ATPase peripheral stalks (26, 27), the catalytic subunit (V1 subunit A) (28), regulatory V1 subunit B (15), and the N-terminal domain of subunit a (28). Recently, Jeffries and Forgac (29) have found that cysteines introduced into the C-terminal domain of subunit H can be cross-linked to subunit F in isolated V1 sectors via a 10-Å cross-linking reagent.In this work, we examine both the subunit-subunit interactions and functional roles of the H-NT and H-CT domains in inhibition of V1-ATPase activity. When expressed in yeast cells lacking subunit H, H-NT can be isolated with cytosolic V1 complexes, but H-CT cannot. We find that both of these domains contribute to inhibition of ATPase activity, but that stable binding to V1 and full inhibition of activity requires both domains. We also find that the H-CT can bind to the cytosolic N-terminal domain of V0 subunit Vph1p (Vph1-NT) in isolation, but does not support tight binding of Vph1-NT to isolated V1 complexes.  相似文献   

15.
A key structural element in the ion translocating F-, A-, and V-ATPases is the peripheral stalk, an assembly of two polypeptides that provides a structural link between the ATPase and ion channel domains. Previously, we have characterized the peripheral stalk forming subunits E and H of the A-ATPase from Thermoplasma acidophilum and demonstrated that the two polypeptides interact to form a stable heterodimer with 1:1 stoichiometry (Kish-Trier, E., Briere, L. K., Dunn, S. D., and Wilkens, S. (2008) J. Mol. Biol. 375, 673–685). To define the domain architecture of the A-ATPase peripheral stalk, we have now generated truncated versions of the E and H subunits and analyzed their ability to bind each other. The data show that the N termini of the subunits form an α-helical coiled-coil, ∼80 residues in length, whereas the C-terminal residues interact to form a globular domain containingα- and β-structure. We find that the isolated C-terminal domain of the E subunit exists as a dimer in solution, consistent with a recent crystal structure of the related Pyrococcus horikoshii A-ATPase E subunit (Lokanath, N. K., Matsuura, Y., Kuroishi, C., Takahashi, N., and Kunishima, N. (2007) J. Mol. Biol. 366, 933–944). However, upon the addition of a peptide comprising the C-terminal 21 residues of the H subunit (or full-length H subunit), dimeric E subunit C-terminal domain dissociates to form a 1:1 heterodimer. NMR spectroscopy was used to show that H subunit C-terminal peptide binds to E subunit C-terminal domain via the terminal α-helices, with little involvement of the β-sheet region. Based on these data, we propose a structural model of the A-ATPase peripheral stalk.The archaeal ATP synthase (A1A0-ATPase),2 along with the related F1F0- and V1V0-ATPases (proton pumping vacuolar ATPases), is a rotary molecular motor (14). The rotary ATPases are bilobular in overall architecture, with one lobe comprising the water-soluble A1, F1, or V1 and the other comprising the membrane-bound A0, F0, or V0 domain, respectively. The subunit composition of the A-ATPase is A3B3DE2FH2 for the A1 and CIKx for the A0. In the A1 domain, the three A and B subunits come together in an alternating fashion to form a hexamer with a hydrophobic inner cavity into which part of the D subunit is inserted. Subunits D and F comprise the central stalk connection to A0, whereas two heterodimeric EH complexes are thought to form the peripheral stalk attachment to A0 seen in electron microscopy reconstructions (5, 6). In the A0 domain (subunits CIKx), the K subunits (proteolipids) form a ring that is linked to the central stalk by the C subunit, whereas the cytoplasmic N-terminal domain of the I subunit probably mediates the binding of the EH peripheral stalks to A0, as suggested for the bacterial A/V-type enzyme (7). Although closer in structure to the proton-pumping V-ATPase, the A-ATPase functions in vivo as an ATP synthase, coupling ion motive force to ATP synthesis, most likely via a similar rotary mechanism as demonstrated for the bacterial A/V- and the vacuolar type enzymes (8, 9). During catalysis, substrate binding occurs sequentially on the three catalytic sites, which are formed predominantly by the A subunits. This is accompanied by conformation changes in the A3B3 hexamer that are linked to the rotation of the embedded D subunit together with the rotor subunits F, C, and the proteolipid ring. Each copy of K contains a lipid-exposed carboxyl residue (Asp or Glu), which is transiently interfaced with the membrane-bound domain of I during rotation, thereby catalyzing ion translocation. The EH peripheral stalks function to stabilize the A3B3 hexamer against the torque generated during rotation of the central stalk. Much work has been accomplished to elucidate the architectural features of the rotational and catalytic domains, especially in the related F- and V-type enzymes. However, the peripheral stalk complexes in the A- and V-type enzymes remain an area open to question. Although the stoichiometry of the peripheral stalks in the A/V-type and the vacuolar type ATPases have recently been resolved to two and three, respectively (6, 10), the overall structure of the peripheral stalk, including the nature of attachment to the A3B3 hexamer and I subunit (called subunit a in the F- and V-ATPase), is not well understood. Some structural information exists in the form of the A-ATPase E subunit C-terminal domain (11), although isolation from its binding partner H may have influenced its conformation.Previously, our lab has characterized the Thermoplasma acidophilum A-ATPase E and H subunits individually and in complex (12). We found that despite their tendency to oligomerize when isolated separately, upon mixing, E and H form a tight heterodimer that was monodisperse and elongated in solution, which is consistent with its role as the peripheral stalk element in the A-ATPase. Here, we have expanded our study of the A-ATPase EH complex through the production of various N- and C-terminal truncation mutants of both binding partners. The data show that the EH complex is comprised of two distinct domains, one that contains both N termini interacting via a coiled-coil and a second that contains both C termini folded in a globular structure containing mixed secondary structure. Consistent with recent crystallographic data for the related A-ATPase from Pyrococcus horikoshii (11), we found that the isolated C-terminal domain of the E subunit exists as a stable homodimer in solution. However, the addition of subunit H or a peptide consisting of the 21 C-terminal residues of the subunit to the dimeric C-terminal domain of subunit E resulted in dissociation of the homodimer with concomitant formation of a 1:1 heterodimer containing the C termini of both polypeptides. This study delineates and characterizes the two domains of the EH complex and will aid in the further exploration of the nature of peripheral stalk attachment and function in the intact A1A0-ATPase.  相似文献   

16.
The vacuolar ATPase (V-ATPase) is a multisubunit complex that carries out ATP-driven proton transport. It is composed of a peripheral V1 domain that hydrolyzes ATP and an integral V0 domain that translocates protons. Subunit a is a 100-kDa integral membrane protein (part of V0) that possesses an N-terminal cytoplasmic domain and a C-terminal hydrophobic domain. Although the C-terminal domain functions in proton transport, the N-terminal domain is critical for intracellular targeting and regulation of V-ATPase assembly. Despite its importance, there is currently no high resolution structure for subunit a of the V-ATPase. Recently, the crystal structure of the N-terminal domain of the related subunit I from the archaebacterium Meiothermus ruber was reported. We have used homology modeling to construct a model of the N-terminal domain of Vph1p, one of two isoforms of subunit a expressed in yeast. To test this model, unique cysteine residues were introduced into a Cys-less form of Vph1p and their accessibility to modification by the sulfhydryl reagent 3-(N-maleimido-propionyl) biocytin (MPB) was determined. In addition, accessibility of introduced cysteine residues to MPB modification was compared in the V1V0 complex and the free V0 domain to identify residues protected from modification by the presence of V1. The results provide an experimental test of the proposed model and have identified regions of the N-terminal domain of subunit a that likely serve as interfacial contact sites with the peripheral V1 domain. The possible significance of these results for in vivo regulation of V-ATPase assembly is discussed.  相似文献   

17.
Two proton pumps, the F-ATPase (ATP synthase, FoF1) and the V-ATPase (endomembrane proton pump), have different physiological functions, but are similar in subunit structure and mechanism. They are composed of a membrane extrinsic (F1 or V1) and a membrane intrinsic (Fo or Vo) sector, and couple catalysis of ATP synthesis or hydrolysis to proton transport by a rotational mechanism. The mechanism of rotation has been extensively studied by kinetic, thermodynamic and physiological approaches. Techniques for observing subunit rotation have been developed. Observations of micron-length actin filaments, or polystyrene or gold beads attached to rotor subunits have been highly informative of the rotational behavior of ATP hydrolysis-driven rotation. Single molecule FRET experiments between fluorescent probes attached to rotor and stator subunits have been used effectively in monitoring proton motive force-driven rotation in the ATP synthesis reaction. By using small gold beads with diameters of 40-60 nm, the E. coli F1 sector was found to rotate at surprisingly high speeds (> 400 rps). This experimental system was used to assess the kinetics and thermodynamics of mutant enzymes. The results revealed that the enzymatic reaction steps and the timing of the domain interactions among the β subunits, or between the β and γ subunits, are coordinated in a manner that lowers the activation energy for all steps and avoids deep energy wells through the rotationally-coupled steady-state reaction. In this review, we focus on the mechanism of steady-state F1-ATPase rotation, which maximizes the coupling efficiency between catalysis and rotation.  相似文献   

18.
The biosynthesis and assembly of the peripheral sector (V1) of the vacuolar protontranslocating adenosine triphosphatase (V-ATPase) was studied in a bovine kidney epithelial cell line. Monolayer cultures of cells were metabolically radiolabeled with Tran 35S-label and the V-ATPase subsequently immunoprecipitated using a monoclonal antibody raised against the bovine brain-coated vesicle proton pump. The V-ATPase immunoprecipitated from the bovine kidney cell line has a subunit composition very similar to that of the bovine brain-coated vesicle proton pump and the V-ATPase prepared from other kidney tissues. Radiolabeling the cells for increasing times showed that the V1 or peripheral portion of the V-ATPase is assembled within 10–15 min; the intact V1V0 complex is also detectable within 10–15 min. Fractionation of the cells into cytosolic and membrane components prior to immunoprecipitation revealed that there is a significant pool of V1 in the cytosol; a similar complex is also found in bovine brain cytosol. Pulse-chase studies suggest that this cytosolic pool is not an obligate precursor for membranebound V1V0 and does not exchange with the membrane V1 population at later times. No qualitative differences in assembly were observed when pulse-chase studies were performed at 15°C or in the presence of brefeldin A. This suggests that assembly of V1V0 is probably completed in the endoplasmic reticulum prior to distribution of the enzyme throughout the cell, with a cytosolic pool of V1 of unknown function existing in parallel with the fully assembled complex. © 1993 Wiley-Liss, Inc.  相似文献   

19.
Summary Differences in the activity and structure of the vacuolar H+-ATPase (V-ATPase, EC 3.6.1.3) were investigated in the C3/CAM intermediate plantKalanchoë blossfeldiana Poellnitz cv. Tom Thumb, with lower or higher expression of CAM, andHordeum vulgare cv. Carina, grown with or without 150 mM NaCl. InK. blossfeldiana ATP-hydrolysis and H+-transport activity were higher with higher expression of CAM than in plants with very weak CAM. This was mainly due to a larger amount of V-ATPase. Statistical analysis of the diameter of intramembrane particles (IMPs) on freeze-fractures of tonoplast vesicles showed that IMPs were larger in tonoplast vesicle preparations ofK. blossfeldiana with strong CAM expression (9.1 nm) than in preparations ofK. blossfeldiana with low CAM expression (7.3 nm). As there is evidence that the majority of IMPs on freeze-fractures of tonoplast vesicles corresponds to the V0 domain of V-ATPase, the higher activity of V-ATPase inK. blossfeldiana with stronger CAM could be a result of additional structural changes in its membrane-integral domain. The higher activity of V-ATPase inK. blossfeldiana with stronger CAM is discussed in relation to the requirement for a higher proton pumping capacity for nocturnal malate accumulation in the vacuole. The ATP-dependent H+-pumping activity inH. vulgare was higher under salt stress than in control plants, while the rates of ATP-hydrolysis and the size of IMPs were not affected by the salt treatment. The data presented here indicate that different mechanisms might increase the transport capacity of V-ATPase to meet the higher requirements of secondary active transport related to CAM expression and adaptation to salt stress.Abbrevations ATP adenosine triphosphate - CAM crassulacean acid metabolism - IMP intramembrane particles - V-ATPase vacuolar proton-translocating adenosine triphosphatase - V0 domain membrane-integral domain of V-ATPase - V1 domain membrane-peripheral domain of V-ATPase Dedicated to Prof. Dr. Eberhard Schnepf on the occasion of his retirement  相似文献   

20.
Synthesis of adenosine triphosphate (ATP) by the F1F0 ATP synthase involves a membrane-embedded rotary engine, the F0 domain, which drives the extra-membranous catalytic F1 domain. The F0 domain consists of subunits a1b2 and a cylindrical rotor assembled from 9–14 α-helical hairpin-shaped c-subunits. According to structural analyses, rotors contain 10 c-subunits in yeast and 14 in chloroplast ATP synthases. We determined the rotor stoichiometry of Ilyobacter tartaricus ATP synthase by atomic force microscopy and cryo-electron microscopy, and show the cylindrical sodium-driven rotor to comprise 11 c-subunits.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号