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 46 毫秒
1.
Cholesterol is the major sterol component of all mammalian plasma membranes. Recent studies have shown that cholesterol inhibits both bacterial (KirBac1.1 and KirBac3.1) and eukaryotic (Kir2.1) inward rectifier K+ (Kir) channels. Lipid-sterol interactions are not enantioselective, and the enantiomer of cholesterol (ent-cholesterol) does not inhibit Kir channel activity, suggesting that inhibition results from direct enantiospecific binding to the channel, and not indirect effects of changes to the bilayer. Furthermore, conservation of the effect of cholesterol among prokaryotic and eukaryotic Kir channels suggests an evolutionary conserved cholesterol-binding pocket, which we aimed to identify. Computational experiments were performed by docking cholesterol to the atomic structures of Kir2.2 (PDB: 3SPI) and KirBac1.1 (PDB: 2WLL) using Autodock 4.2. Poses were assessed to ensure biologically relevant orientation and then clustered according to location and orientation. The stability of cholesterol in each of these poses was then confirmed by molecular dynamics simulations. Finally, mutation of key residues (S95H and I171L) in this putative binding pocket found within the transmembrane domain of Kir2.1 channels were shown to lead to a loss of inhibition by cholesterol. Together, these data provide support for this location as a biologically relevant pocket.  相似文献   

2.
We have performed discontinuous molecular dynamic simulations of the assembly and folding kinetics of a tetrameric β-sheet complex that contains four identical four-stranded antiparallel β-sheet peptides. The potential used in the simulation is a hybrid Go-type potential characterized by the bias gap parameter g, an artificial measure of a model protein's preference for its native state, and the intermolecular contact parameter η, which measures the ratio of intermolecular to intramolecular native attractions. The formation of the β-sheet complex and its equilibrium properties strongly depend on the size of the intermolecular contact parameter η. The ordered β-sheet complex in the folded state and nonaligned β-sheets or tangled chains in the misfolded state are distinguished by measuring the squared radius of gyration and the fraction of native contacts Q. The folding yield for the folded state is high at intermediate values of η, but is low at both small and large values of η. The folded state at small η is liquid-like, but is solid-like at both intermediate and large η. The misfolded state at small η contains nonaligned β-sheets and tangled chains with poor secondary structure at large η. Various folding pathways via dimeric and trimeric intermediates are observed, depending on η. Comparison with experimental results on protein aggregation indicates that intermediate η values are most appropriate for modeling fibril formation and small η values are most appropriate for modeling the formation of amorphous aggregates.  相似文献   

3.
Staphylococcus aureus expresses numerous virulence factors that aid in immune evasion. The four-domain staphylococcal immunoglobulin binding (Sbi) protein interacts with complement component 3 (C3) and its thioester domain (C3d)-containing fragments. Recent structural data suggested two possible modes of binding of Sbi domain IV (Sbi-IV) to C3d, but the physiological binding mode remains unclear. We used a computational approach to provide insight into the C3d-Sbi-IV interaction. Molecular dynamics (MD) simulations showed that the first binding mode (PDB code 2WY8) is more robust than the second (PDB code 2WY7), with more persistent polar and nonpolar interactions, as well as conserved interfacial solvent accessible surface area. Brownian dynamics and steered MD simulations revealed that the first binding mode has faster association kinetics and maintains more stable intermolecular interactions compared to the second binding mode. In light of available experimental and structural data, our data confirm that the first binding mode represents Sbi-IV interaction with C3d (and C3) in a physiological context. Although the second binding mode is inherently less stable, we suggest a possible physiological role. Both binding sites may serve as a template for structure-based design of novel complement therapeutics.  相似文献   

4.
Septins comprise a family of proteins involved in a variety of cellular processes and related to several human pathologies. They are constituted by three structural domains: the N- and C-terminal domains, highly variable in length and composition, and the central domain, involved in the guanine nucleotide (GTP) binding. Thirteen different human septins are known to form heterogeneous complexes or homofilaments, which are stabilized by specific interactions between the different interfaces present in the domains. In this work, we have investigated by in-solution small-angle x-ray scattering the structural and thermodynamic properties of a human septin 3 construct, SEPT3-GC, which contains both of both interfaces (G and NC) responsible for septin-septin interactions. In order to shed light on the role of these interactions, small-angle x-ray scattering measurements were performed in a wide range of temperatures, from 2 up to 56°C, both with and without a nonhydrolysable form of GTP (GTPγS). The acquired data show a temperature-dependent coexistence of monomers, dimers, and higher-order aggregates that were analyzed using a global fitting approach, taking into account the crystallographic structure of the recently reported SEPT3 dimer, PDB:3SOP. As a result, the enthalpy, entropy, and heat capacity variations that control the dimer-monomer dissociation equilibrium in solution were derived and GTPγS was detected to increase the enthalpic stability of the dimeric species. Moreover, a temperature increase was observed to induce dissociation of SEPT3-GC dimers into monomers just preceding their reassembling into amyloid aggregates, as revealed by the Thioflavin-T fluorescence assays.  相似文献   

5.
Sup35p of Saccharomyces cerevisiae can form the [PSI+] prion, an infectious amyloid in which the protein is largely inactive. The part of Sup35p that forms the amyloid is the region normally involved in control of mRNA turnover. The formation of [PSI+] by Sup35p’s from other yeasts has been interpreted to imply that the prion-forming ability of Sup35p is conserved in evolution, and thus of survival/fitness/evolutionary value to these organisms. We surveyed a larger number of yeast and fungal species by the same criteria as used previously and find that the Sup35p from many species cannot form prions. [PSI+] could be formed by the Sup35p from Candida albicans, Candida maltosa, Debaromyces hansenii, and Kluyveromyces lactis, but orders of magnitude less often than the S. cerevisiae Sup35p converts to the prion form. The Sup35s from Schizosaccharomyces pombe and Ashbya gossypii clearly do not form [PSI+]. We were also unable to detect [PSI+] formation by the Sup35ps from Aspergillus nidulans, Aspergillus fumigatus, Magnaporthe grisea, Ustilago maydis, or Cryptococcus neoformans. Each of two C. albicans SUP35 alleles can form [PSI+], but transmission from one to the other is partially blocked. These results suggest that the prion-forming ability of Sup35p is not a conserved trait, but is an occasional deleterious side effect of a protein domain conserved for another function.  相似文献   

6.
We show by whole genome sequence analysis that loss of RNase H2 activity increases loss of heterozygosity (LOH) in Saccharomyces cerevisiae diploid strains harboring the pol2-M644G allele encoding a mutant version of DNA polymerase ε that increases ribonucleotide incorporation. This led us to analyze the effects of loss of RNase H2 on LOH and on nonallelic homologous recombination (NAHR) in mutant diploid strains with deletions of genes encoding RNase H2 subunits (rnh201Δ, rnh202Δ, and rnh203Δ), topoisomerase 1 (TOP1Δ), and/or carrying mutant alleles of DNA polymerases ε, α, and δ. We observed an ∼7-fold elevation of the LOH rate in RNase H2 mutants encoding wild-type DNA polymerases. Strains carrying the pol2-M644G allele displayed a 7-fold elevation in the LOH rate, and synergistic 23-fold elevation in combination with rnh201Δ. In comparison, strains carrying the pol2-M644L mutation that decreases ribonucleotide incorporation displayed lower LOH rates. The LOH rate was not elevated in strains carrying the pol1-L868M or pol3-L612M alleles that result in increased incorporation of ribonucleotides during DNA synthesis by polymerases α and δ, respectively. A similar trend was observed in an NAHR assay, albeit with smaller phenotypic differentials. The ribonucleotide-mediated increases in the LOH and NAHR rates were strongly dependent on TOP1. These data add to recent reports on the asymmetric mutagenicity of ribonucleotides caused by topoisomerase 1 processing of ribonucleotides incorporated during DNA replication.  相似文献   

7.

Rationale

To prevent or combat infection, increasing the effectiveness of the immune response is highly desirable, especially in case of compromised immune system function. However, immunostimulatory therapies are scarce, expensive, and often have unwanted side-effects. β-glucans have been shown to exert immunostimulatory effects in vitro and in vivo in experimental animal models. Oral β-glucan is inexpensive and well-tolerated, and therefore may represent a promising immunostimulatory compound for human use.

Methods

We performed a randomized open-label intervention pilot-study in 15 healthy male volunteers. Subjects were randomized to either the β -glucan (n = 10) or the control group (n = 5). Subjects in the β-glucan group ingested β-glucan 1000 mg once daily for 7 days. Blood was sampled at various time-points to determine β-glucan serum levels, perform ex vivo stimulation of leukocytes, and analyze microbicidal activity.

Results

β-glucan was barely detectable in serum of volunteers at all time-points. Furthermore, neither cytokine production nor microbicidal activity of leukocytes were affected by orally administered β-glucan.

Conclusion

The present study does not support the use of oral β-glucan to enhance innate immune responses in humans.

Trial Registration

ClinicalTrials.gov NCT01727895  相似文献   

8.
Genetic screens in Drosophila melanogaster and other organisms have been pursued to filter the genome for genetic functions important for memory formation. Such screens have employed primarily chemical or transposon-mediated mutagenesis and have identified numerous mutants including classical memory mutants, dunce and rutabaga. Here, we report the results of a large screen using panneuronal RNAi expression to identify additional genes critical for memory formation. We identified >500 genes that compromise memory when inhibited (low hits), either by disrupting the development and normal function of the adult animal or by participating in the neurophysiological mechanisms underlying memory formation. We also identified >40 genes that enhance memory when inhibited (high hits). The dunce gene was identified as one of the low hits and further experiments were performed to map the effects of the dunce RNAi to the α/β and γ mushroom body neurons. Additional behavioral experiments suggest that dunce knockdown in the mushroom body neurons impairs memory without significantly affecting acquisition. We also characterized one high hit, sickie, to show that RNAi knockdown of this gene enhances memory through effects in dopaminergic neurons without apparent effects on acquisition. These studies further our understanding of two genes involved in memory formation, provide a valuable list of genes that impair memory that may be important for understanding the neurophysiology of memory or neurodevelopmental disorders, and offer a new resource of memory suppressor genes that will aid in understanding restraint mechanisms employed by the brain to optimize resources.  相似文献   

9.
DNA polymerases (Pols) ε and δ perform the bulk of yeast leading- and lagging-strand DNA synthesis. Both Pols possess intrinsic proofreading exonucleases that edit errors during polymerization. Rare errors that elude proofreading are extended into duplex DNA and excised by the mismatch repair (MMR) system. Strains that lack Pol proofreading or MMR exhibit a 10- to 100-fold increase in spontaneous mutation rate (mutator phenotype), and inactivation of both Pol δ proofreading (pol3-01) and MMR is lethal due to replication error-induced extinction (EEX). It is unclear whether a similar synthetic lethal relationship exists between defects in Pol ε proofreading (pol2-4) and MMR. Using a plasmid-shuffling strategy in haploid Saccharomyces cerevisiae, we observed synthetic lethality of pol2-4 with alleles that completely abrogate MMR (msh2Δ, mlh1Δ, msh3Δ msh6Δ, or pms1Δ mlh3Δ) but not with partial MMR loss (msh3Δ, msh6Δ, pms1Δ, or mlh3Δ), indicating that high levels of unrepaired Pol ε errors drive extinction. However, variants that escape this error-induced extinction (eex mutants) frequently emerged. Five percent of pol2-4 msh2Δ eex mutants encoded second-site changes in Pol ε that reduced the pol2-4 mutator phenotype between 3- and 23-fold. The remaining eex alleles were extragenic to pol2-4. The locations of antimutator amino-acid changes in Pol ε and their effects on mutation spectra suggest multiple mechanisms of mutator suppression. Our data indicate that unrepaired leading- and lagging-strand polymerase errors drive extinction within a few cell divisions and suggest that there are polymerase-specific pathways of mutator suppression. The prevalence of suppressors extragenic to the Pol ε gene suggests that factors in addition to proofreading and MMR influence leading-strand DNA replication fidelity.  相似文献   

10.
11.
Understanding which peptides and proteins have the potential to undergo amyloid formation and what driving forces are responsible for amyloid-like fiber formation and stabilization remains limited. This is mainly because proteins that can undergo structural changes, which lead to amyloid formation, are quite diverse and share no obvious sequence or structural homology, despite the structural similarity found in the fibrils. To address these issues, a novel approach based on recursive feature selection and feed-forward neural networks was undertaken to identify key features highly correlated with the self-assembly problem. This approach allowed the identification of seven physicochemical and biochemical properties of the amino acids highly associated with the self-assembly of peptides and proteins into amyloid-like fibrils (normalized frequency of β-sheet, normalized frequency of β-sheet from LG, weights for β-sheet at the window position of 1, isoelectric point, atom-based hydrophobic moment, helix termination parameter at position j+1 and ΔG° values for peptides extrapolated in 0 M urea). Moreover, these features enabled the development of a new predictor (available at http://cran.r-project.org/web/packages/appnn/index.html) capable of accurately and reliably predicting the amyloidogenic propensity from the polypeptide sequence alone with a prediction accuracy of 84.9 % against an external validation dataset of sequences with experimental in vitro, evidence of amyloid formation.  相似文献   

12.
The release of GDP from GTPases signals the initiation of a GTPase cycle, where the association of GTP triggers conformational changes promoting binding of downstream effector molecules. Studies have implicated the nucleotide-binding G5 loop to be involved in the GDP release mechanism. For example, biophysical studies on both the eukaryotic Gα proteins and the GTPase domain (NFeoB) of prokaryotic FeoB proteins have revealed conformational changes in the G5 loop that accompany nucleotide binding and release. However, it is unclear whether this conformational change in the G5 loop is a prerequisite for GDP release, or, alternatively, the movement is a consequence of release. To gain additional insight into the sequence of events leading to GDP release, we have created a chimeric protein comprised of Escherichia coli NFeoB and the G5 loop from the human Giα1 protein. The protein chimera retains GTPase activity at a similar level to wild-type NFeoB, and structural analyses of the nucleotide-free and GDP-bound proteins show that the G5 loop adopts conformations analogous to that of the human nucleotide-bound Giα1 protein in both states. Interestingly, isothermal titration calorimetry and stopped-flow kinetic analyses reveal uncoupled nucleotide affinity and release rates, supporting a model where G5 loop movement promotes nucleotide release.The hydrolysis of guanosine triphosphate (GTP) by GTPases, such as the oncoprotein p21 Ras and heterotrimeric Gα proteins, is a critical regulatory activity for cell growth and proliferation (1). Aberrant GTPases are consequently often implicated in tumorigenesis, developmental disorders, and metabolic diseases (2). Critical for the initiation of a GTPase cycle is the release of guanosine diphosphate (GDP), which allows GTP to bind and switch the protein from an inactive to an active conformation. The GTP is subsequently hydrolyzed to GDP and inorganic phosphate, returning the GTPase to an inactive conformation (3).Given that the release of GDP is the fundamental step in the initiation of a GTPase cycle, the detailed mechanism by which it is released has been under intense scrutiny. Studies using double electron-electron resonance, deuterium-exchange, Rosetta energy analysis, and electron paramagnetic resonance, have shown that the mechanism involves conformational changes in the nucleotide-coordinating G5 loop, one of five nucleotide recognition motifs (4, 5, 6, 7, 8, 9, 10, 11). Structural studies of eukaryotic Gα proteins and the intracellular TEES-type GTPase domain of the prokaryotic iron transporter FeoB (NFeoB) have also illustrated distinct conformations of the G5 loop, depending on the nucleotide-bound state (9, 12).Recently, we reported mutational studies of the G5 loop of Escherichia coli NFeoB, which illustrated a correlation between the sequence composition of the loop and the intrinsic GDP release rate (13). However, despite these observations, it is unclear whether the observed conformational changes in the G5 loop are a prerequisite for GDP release, or if the movement is a consequence of GDP release. To address this fundamental question, in this study we have used a combination of protein engineering and biophysical methods.Initially, to assess the relevance of conformational flexibility in the G5 loop, we aimed to create a protein chimera combining sequence and structural characteristics of both fast and slow GDP-releasing GTPases. We thus engineered a protein chimera using E. coli NFeoB as the scaffold (a protein with fast intrinsic GDP release) and substituted the G5 loop with that of a slow GDP-releasing protein (the human Giα1 protein; Gene ID 2770; Fig. 1 A (5)). GTP hydrolysis assays comparing wild-type (wt) NFeoB (wtNFeoB) and the protein chimera (ChiNFeoB) validated the integrity of the GTPase activities of both proteins (kcat = 0.46 and 0.36 min−1, respectively). To further assess the ChiNFeoB protein, we determined its crystal structure at 2.2 Å resolution (see Table S1 in the Supporting Material). The ChiNFeoB structure contains two molecules in the asymmetric unit, with molecule A bound to GDP. They are essentially identical to the nucleotide-bound wtNFeoB structure (root-mean-square deviation of 1.2 Å over 226 Cα atoms; Fig. 2).Open in a separate windowFigure 1Chimera model and structural comparison. (A) Illustration highlighting the chimera sequence change. (Orange) Sequence of the extended G5 loop from Giα1, which replaced the NFeoB sequence (gray). (B–F) Structural comparison of the G5 loop between (B) WT apo (PDB:3HYR) and nucleotide-bound (PDB:3HYT) NFeoB structures. (C) NFeoB nucleotide-bound and Giα1 (PDB:2ZJZ). (D) Nucleotide-bound NFeoB and chimera (Chi_GDP). (E) Nucleotide-bound chimera and Giα1. (F) Nucleotide-free (Chi_apo) and bound chimera protein. (G) Overview of the nucleotide binding site and structural overlay of chimera and Giα1 structures. To see this figure in color, go online.Open in a separate windowFigure 2Superimposition of nucleotide-bound NFeoB and chimera protein, with thermodynamic parameters. To see this figure in color, go online.However, the ChiNFeoB structure, when compared to the wtNFeoB structure, revealed an alteration in the conformation of the G5 loop, showing an extra turn on the N-terminal end of the α6 helix. This is structurally distinct from the wtFeoB protein, but with a conformation similar to that of the Giα1 protein (PDB:2ZJZ; Fig. 1, B–F). As in the crystal structures of wtNFeoB and Giα1, ChiNFeoB residues implicated in coordination of the nucleotide base maintain their positions in the G5 loop relative to GDP. In particular, residues Ala150 and Thr151 (NFeoB numbering, the asterisk indicates Giα1 chimera residue) are involved in electrostatic interactions with the nucleotide base moiety, analogous to the structures of both wtNFeoB and Giα1 (Fig. 1 G). Serendipitously, the second molecule in the asymmetric unit of ChiNFeoB (molecule B) was present in the nucleotide-free state. The two molecules (GDP-bound and nucleotide-free) are nearly identical (the superposition of molecules A and B yields a root-mean-square deviation of 0.36 Å over 229 Cα atoms), with the G5 loop adopting a nearly indistinguishable conformation compared to that of the GDP-bound molecule A (Fig. 1 F).Importantly, this conformation is independent of the crystallographic packing, inasmuch as the loop is not involved in any crystal contacts. In contrast, the structures of nucleotide-bound and nucleotide-free wtNFeoB illustrated a large conformational change in the G5 loop (Fig. 1 B). Hence, the substitution in the chimera extends the secondary structure of the α6 helix, and as hypothesized, the engineered ChiNFeoB protein has a G5 loop structure that is more conformationally stable than that of wtNFeoB.We subsequently measured the affinity of the ChiNFeoB protein for GDP using isothermal titration calorimetry (ITC). Nonlinear regression was used to attain the thermodynamic parameters (including the GDP binding affinity, Ka; the corresponding dissociation constant (Kd) was calculated from the equation Kd = 1/Ka). Interestingly, these measurements revealed the ChiNFeoB protein to have an almost 10-fold reduced affinity for GDP (82 vs. 9 μM measured for the WT protein; Fig. 2). In contrast, in a recent alanine scanning mutagenesis study of the G5 loop we observed a fivefold increase in affinity for GDP in a Ser150Ala mutant (2 μM) (14). This mutant protein has a coordination environment for the GDP base analogous to that of the ChiNFeoB protein (Fig. 1 A), indicating that it is not the presence of an alanine at position 150 that causes the reduced GDP affinity observed for the chimera protein. Instead, the analysis by ITC and comparison with previous mutagenesis studies indicates that the GDP binding site is less accessible in the ChiNFeoB protein, likely due to the introduction of conformational rigidity that accompanies the extension of secondary structural elements within the loop (Fig. 1 D).To further evaluate the functional characteristics of the chimera protein, we used stopped-flow fluorescence assays to determine the rate of nucleotide dissociation (koff) and association (kon) for the ChiNFeoB protein. The association rate for the GTP analog mant-GMPPNP was determined from the slope of a linear plot of protein concentration versus the observed association constant (kobs). The kon for the chimera was determined to be 3.20 μM−1 min−1 (Supporting Material), the dissociation rate (koff) of GDP for the chimera was determined to be 16.6 s−1 (vs. 144 s−1 for wtNFeoB;
DesignationmGMPPNPmGDP
Proteinkona (μM−1 min−1)koffb (min−1)Kdc (μM)kond (μM−1 min−1)koffe (s−1)
NFeoB8.1 ± 0.178.6 ± 1.69.715.9144.7 ± 2.0
Chimera3.2 ± 0.1208.2 ± 1.365.10.216.61 ± 0.50
Open in a separate windowAll values are the average of three or more stopped-flow experiments with each experiment consisting of five or more replicates.akon was determined from the slope of the linear plot formed by kobs at protein concentrations between 1.25 and 40 μM.bkoff was determined from the y-intercept of the linear plot.cKd was determined from the ratio of koff to kon.dkon was determined from the ratio of koff (mGDP) to Kd (GDP; ITC).emGDP dissociation rates (koff) were determined by fitting a single exponential function to stopped-flow data.We have previously observed a consistent correlation between nucleotide affinity and release rates (e.g., high affinity, slow release), and the uncoupling of this relationship, observed in this study, provides clues to the mechanism of the nucleotide release in GTPases. As observed in our structural analysis, the extension of the α6 helix in the chimera protein generates a shorter G5 loop that is more stable in the nucleotide-coordinating conformation, a conformation retained in both the GDP-bound and the apo states of the protein. Because the nucleotide pocket remains capped, it is likely to be less accessible for nucleotide binding, providing a rationale for its reduced GDP affinity (Fig. 2) and on-rate (Fig. 1 B) in particular, likely plays a significant role in the observed rapid intrinsic GDP release mechanism (12, 15). Future studies generating a reciprocal chimera, using the Giα1 protein as a scaffold and the FeoB G5 motif insert, could provide further support for these results.In summary, our combined results support a model where G5 loop movement precedes GDP release, and illustrates that loop movement can act to catalyze both intrinsic and coupled nucleotide release.  相似文献   

13.
RCSB Protein Data bank: Tools for visualizing and understanding biological macromolecules in 3D     
《Protein science : a publication of the Protein Society》2022,31(12)
Now in its 52nd year of continuous operations, the Protein Data Bank (PDB) is the premiere open‐access global archive housing three‐dimensional (3D) biomolecular structure data. It is jointly managed by the Worldwide Protein Data Bank (wwPDB) partnership. The Research Collaboratory for Structural Bioinformatics Protein Data Bank (RCSB PDB) is funded by the National Science Foundation, National Institutes of Health, and US Department of Energy and serves as the US data center for the wwPDB. RCSB PDB is also responsible for the security of PDB data in its role as wwPDB‐designated Archive Keeper. Every year, RCSB PDB serves tens of thousands of depositors of 3D macromolecular structure data (coming from macromolecular crystallography, nuclear magnetic resonance spectroscopy, electron microscopy, and micro‐electron diffraction). The RCSB PDB research‐focused web portal (RCSB.org) makes PDB data available at no charge and without usage restrictions to many millions of PDB data consumers around the world. The RCSB PDB training, outreach, and education web portal (PDB101.RCSB.org) serves nearly 700 K educators, students, and members of the public worldwide. This invited Tools Issue contribution describes how RCSB PDB (i) is organized; (ii) works with wwPDB partners to process new depositions; (iii) serves as the wwPDB‐designated Archive Keeper; (iv) enables exploration and 3D visualization of PDB data via RCSB.org; and (v) supports training, outreach, and education via PDB101.RCSB.org. New tools and features at RCSB.org are presented using examples drawn from high‐resolution structural studies of proteins relevant to treatment of human cancers by targeting immune checkpoints.  相似文献   

14.
The contribution of αβ-tubulin curvature to microtubule dynamics     
Gary J. Brouhard  Luke M. Rice 《The Journal of cell biology》2014,207(3):323-334
Microtubules are dynamic polymers of αβ-tubulin that form diverse cellular structures, such as the mitotic spindle for cell division, the backbone of neurons, and axonemes. To control the architecture of microtubule networks, microtubule-associated proteins (MAPs) and motor proteins regulate microtubule growth, shrinkage, and the transitions between these states. Recent evidence shows that many MAPs exert their effects by selectively binding to distinct conformations of polymerized or unpolymerized αβ-tubulin. The ability of αβ-tubulin to adopt distinct conformations contributes to the intrinsic polymerization dynamics of microtubules. αβ-Tubulin conformation is a fundamental property that MAPs monitor and control to build proper microtubule networks.Microtubules are polar polymers formed from αβ-tubulin heterodimers. These tubulin subunits associate head-to-tail to form protofilaments, and typically 13 protofilaments are associated side-by-side to form the hollow cylindrical microtubule. Most microtubules emanate from microtubule organizing centers, in which their minus ends are embedded. GTP-tubulin associates with the fast-growing plus ends as the microtubules radiate to explore the cell interior (see Box).

The cycle of microtubule polymerization.

Fig. 1). The addition of a new subunit completes the active site for GTP hydrolysis, and consequently most of the body of the microtubule contains GDP-bound αβ-tubulin. The GDP lattice is unstable but protected from depolymerization by a stabilizing “GTP cap,” an extended region of newly added GTP- or GDP.Pi-bound αβ-tubulin. The precise nature of the microtubule end structure and the size and composition of the cap are a matter of debate. Loss of the stabilizing cap leads to rapid depolymerization, which is characterized by an apparent peeling of protofilaments. “Catastrophe” denotes the switch from growth to shrinkage, and “rescue” denotes the switch from shrinkage to growth.Open in a separate windowFigure 1.Three structures of GTP-bound αβ-tubulin adopt similar curved conformations. Different αβ-tubulin structures were superimposed using α-tubulin as a reference, and oligomers were generated by assuming that the spatial relationship between α- and β-tubulin within a heterodimer is identical to the relationship between heterodimers. Curvature is calculated from the rotational component of the transformation required to superimpose the α-tubulin chain onto the β-tubulin chain of the same heterodimer. All of the GTP-bound structures (Rb3 complex, Protein Data Bank [PDB] accession no. 3RYH [magenta]; DARPin complex, PDB accession no. 4DRX [green]; TOG1 complex, PDB accession no. 4FFB [blue]) show between 10° and 13° of curvature, which is very similar to the curvature observed in GDP-bound structures (see inset, where the αβ-tubulins from a GDP-bound stathmin complex [PDB accession no. 1SA0] are shown in yellow and orange). A straight protofilament (putty and dark red color, PDB accession no. 1JFF) and a partially straightened assembly (tan) from GMPCPP ribbons are shown for reference.Unlike actin filaments, which grow steadily, microtubules frequently switch between phases of growth and shrinkage. This hallmark property of microtubules, known as “dynamic instability” (Mitchison and Kirschner, 1984), allows the microtubule cytoskeleton to be remodeled rapidly over the course of the cell cycle. “Catastrophes” are GTPase-dependent transitions from growing to shrinking, whereas “rescues” are transitions from shrinking to growing. Numerous microtubule-associated proteins (MAPs) regulate microtubule polymerization dynamics. Discovering how cells regulate and harness dynamic instability is a fundamental challenge in cell biology.A recent accumulation of structural, biochemical, and in vitro reconstitution data has advanced the understanding of dynamic instability and the MAPs that control it. Fresh structural data have provided insight into the process of microtubule assembly and defined how some MAPs recognize αβ-tubulin in and out of the microtubule. In vitro reconstitution experiments are reshaping the understanding of catastrophe and also providing quantitative insight into the mechanism of MAPs. Here, we review this progress, paying special attention to the emerging theme of interactions that are selective for different conformations of αβ-tubulin, both inside and outside the microtubule lattice. We argue for the central importance of recognizing these distinct conformations in the control of microtubule dynamics by MAPs and hence in the construction of a functional microtubule cytoskeleton by cells.

Tubulin dimers and their curvatures

It was clear in early EM studies that αβ-tubulin could form a diversity of polymers (Kirschner et al., 1974). In particular, the first cryo-EM of dynamic microtubules (Mandelkow et al., 1991) revealed significant differences in the appearance of growing and shrinking microtubule ends. Growing microtubule ends had straight protofilaments and were tapered, with uneven protofilament lengths, whereas shrinking microtubule ends had curved protofilaments that peeled outward and lost their lateral contacts. These and other data established the canonical model that GTP-tubulin is “straight” but GDP-tubulin is “curved” (Melki et al., 1989). The idea that GTP binding straightened αβ-tubulin into a microtubule-compatible conformation before polymerization was appealing because it provided a structural rationale for why microtubule assembly required GTP and how GTP hydrolysis could lead to catastrophe. A subsequent cryo-EM study (Chrétien et al., 1995), however, revealed that growing microtubules often tapered and curved gently outward without losing their lateral contacts. These data suggested that GTP-tubulin might not be fully straight at the time of its incorporation into the microtubule lattice, an observation that set the stage for a still-active debate on the structure of GTP-tubulin and of microtubule ends.The atomic details of “straight” and “curved” became apparent when the first structures of αβ-tubulin were solved. The straight conformation of αβ-tubulin was determined from cryo-electron crystallographic studies of Zn-induced αβ-tubulin sheets (Nogales et al., 1998). The structure showed linear head-to-tail stacking of αβ-tubulin along the protofilament, both within and between αβ-tubulin heterodimers. The curved conformation of αβ-tubulin was determined from x-ray crystallographic studies of a complex between αβ-tubulin and Rb3 (Gigant et al., 2000; Ravelli et al., 2004), a microtubule-destabilizing factor in the Op18/stathmin family (Belmont and Mitchison, 1996). In this complex, the individual α- and β-tubulin chains adopted a characteristic conformation distinct from their straight one. Longitudinal interactions also differed from those in the straight conformation (Fig. 1): within and between the heterodimers, successive α- and β-tubulin chains were related by an ∼12° rotation. A chain of these curved αβ-tubulins generates an arc with a radius of curvature resembling that of the peeling protofilaments at shrinking microtubule ends (Gigant et al., 2000; Steinmetz et al., 2000).Straight and curved are not the only two conformations, however. A cryo-EM study of αβ-tubulin helical ribbons trapped using guanylyl 5′-α,β-methylenediphosphonate (GMPCPP), a slowly hydrolyzable analogue of GTP, provided a molecular view of a possible microtubule assembly intermediate (Wang and Nogales, 2005). In these ribbons, GMPCPP-bound αβ-tubulin adopted a conformation roughly halfway (∼5° rotation) between the straight and curved conformations. These partially curved αβ-tubulin heterodimers formed two types of lateral bonds, only one of which resembled those in the microtubule. This structure suggested that at least some αβ-tubulin straightening occurs during polymerization.Until recently, structural information about the conformation of unpolymerized GTP-bound αβ-tubulin was notably lacking. Three recent crystal structures (Nawrotek et al., 2011; Ayaz et al., 2012; Pecqueur et al., 2012) have now provided remarkably similar views of this previously elusive species. In all three structures, GTP-bound αβ-tubulin adopts a fully curved conformation, with its α- and β-tubulin subunits related by ∼12° of rotation (Fig. 1). This curvature is not consistent with models in which GTP binding straightens unpolymerized αβ-tubulin. In each of the structures, αβ-tubulin is bound to another protein, stathmin/Rb3 (Ozon et al., 1997), a designed ankyrin repeat protein (DARPin; Pecqueur et al., 2012), as well as a TOG domain from the Stu2/XMAP215 family of microtubule polymerases (Gard and Kirschner, 1987; Wang and Huffaker, 1997). Biochemical experiments have failed to detect GTP-induced straightening of αβ-tubulin, arguing against the possibility that these unrelated binding partners forced GTP-tubulin to adopt the curved conformation. For example, the affinity of stathmin–tubulin interactions is the same for GTP-tubulin and GDP-tubulin (Honnappa et al., 2003). Similarly, five small molecule ligands that target the colchicine binding site and are predicted to bind only curved αβ-tubulin have equivalent affinity for GTP-tubulin, GDP-tubulin, and αβ-tubulin in the stathmin complex (Barbier et al., 2010). Likewise, a TOG domain from Stu2p binds to GTP- and GDP-tubulin with comparable affinity (Ayaz et al., 2012). Finally, DARPin binds equally well to GTP- and GDP-tubulin even though it contacts a structural element that is positioned differently in the straight and curved conformations (Pecqueur et al., 2012). Taken together with early biochemical experiments (Manuel Andreu et al., 1989; Shearwin et al., 1994), these new data strongly support a model in which unpolymerized αβ-tubulin is curved whether it is bound to GTP or to GDP (Buey et al., 2006; Rice et al., 2008; Nawrotek et al., 2011). According to this model, the curved-to-straight transition occurs during the polymerization process, not before. We discuss some implications of this new view at the end of the following section.

Conformation and dynamic instability

How does GTP hydrolysis destabilize the microtubule lattice and trigger catastrophe? A recent structural study has compared high-resolution cryo-EM reconstructions of GMPCPP microtubules and GDP microtubules to provide some answers to this question (Alushin et al., 2014). The structures show that GTP hydrolysis induces a compaction at the longitudinal interface between dimers, immediately above the exchangeable nucleotide-binding site. This compaction is accompanied by conformational changes in α-tubulin. In contrast, lateral contacts between tubulins were essentially unchanged in the different nucleotide states. These observations suggest that GTP hydrolysis introduces strain into the lattice, but how this strain affects the strength of longitudinal and lateral bonds to destabilize the microtubule remains unknown. The GMPCPP and GDP microtubules also show distinct arrangements of elements that bind to MAPs, which suggests a structural mechanism some MAPs could use to distinguish GTP lattices from GDP lattices (discussed later).In parallel with these structural advances, in vitro reconstitutions (Gardner et al., 2011b) have undermined the textbook view about the kinetics of catastrophe. The seminal measurements of catastrophe frequency (Walker et al., 1988, 1991) assumed that catastrophe occurred with the same probability on newly formed and old microtubules. In other words, the analysis implied that catastrophe was a first-order, single-step process. Although subsequent experiments (e.g., Odde et al., 1995; Janson et al., 2003) indicated that catastrophe involved multiple steps, the first-order view of catastrophe was widely adopted (Howard, 2001; Phillips et al., 2008). Recent experiments using a single-molecule assay for microtubule growth (Gell et al., 2010) have now shown definitively that catastrophe is not a single-step process; rather, newly formed microtubules undergo catastrophe less frequently than older ones (Gardner et al., 2011b). “Age-dependent” catastrophe implies that the stabilizing structure at the end of growing microtubules is evolving to become less effective. The timescale of this evolution is long compared with the kinetics of αβ-tubulin association (Gardner et al., 2011a). Thus, the ageing process probably reports on one or more structural properties of the microtubule end, such as the presence of “defects” in the lattice (Gardner et al., 2011b) or possibly increased tapering of microtubule ends (Coombes et al., 2013).It now seems clear that changes in the curvature of αβ-tubulin during microtubule polymerization are fundamental to microtubule dynamics and the regulatory activities of MAPs. Having straight conformations of αβ-tubulin only occur appreciably in the microtubule lattice provides a simple structural mechanism by which MAPs can discriminate unpolymerized from polymerized αβ-tubulin. Biochemical properties that define microtubule dynamics, like the strength of lateral and longitudinal contacts and the rate of GTP hydrolysis, may differ for curved, straight, and intermediate conformations of αβ-tubulin; e.g., curved forms probably bind microtubule ends less tightly than straight forms. By regulating when and where these different conformations occur, MAPs can tune microtubule dynamics. More speculatively, the complex biochemistry associated with different conformations of αβ-tubulin may contribute to the aging of microtubule ends, which leads to catastrophe. Understanding the connections between αβ-tubulin conformation, biochemistry, and polymerization dynamics is a major challenge for the future. Expanding the current mathematical models (Bowne-Anderson et al., 2013) and computational models (VanBuren et al., 2005; Margolin et al., 2012) of microtubule dynamics to incorporate these new findings about αβ-tubulin structure and age-dependent catastrophe may yield significant insights. In the following sections, we will examine recent studies that demonstrate how MAPs use selective interactions with distinct conformations of αβ-tubulin to control microtubule dynamics and thereby the physiology of the microtubule cytoskeleton.

Microtubule depolymerases stabilize curved conformations of tubulin

Perhaps the first direct evidence that MAPs might control the conformation of αβ-tubulin came from studies of microtubule depolymerases, which are proteins that promote, accelerate, or induce the depolymerization of microtubules (Howard and Hyman, 2007). Cells use microtubule depolymerases to maintain local control of microtubule catastrophe. Early electron microscopy studies of two unrelated depolymerases, Op18/stathmin and the kinesin-13 Xkcm1, showed that these proteins were able to induce/stabilize the curved conformation of αβ-tubulin and/or curved protofilaments (Desai et al., 1999; Gigant et al., 2000; Steinmetz et al., 2000). Depolymerases are also referred to as “catastrophe factors” because they trigger catastrophes in dynamic microtubules. The localized control of catastrophe is the essential function of depolymerases in cell physiology.The microtubule depolymerase stathmin is inactivated around chromosomes and at the leading edge of migrating cells (Niethammer et al., 2004), creating a gradient of depolymerase activity in these zones. Proteins in the Op18/stathmin family form a tight complex with two curved tubulin dimers (Fig. 2 A). Op18/stathmin proteins have been critical for the crystallization of tubulin (Ravelli et al., 2004; Gigant et al., 2005; Prota et al., 2013) and for biochemical studies of tubulin conformation. Although stathmins are frequently described as tubulin-sequestering proteins, the effect they have on microtubule catastrophe frequencies in vitro is much stronger than would be predicted from the simple sequestration of tubulin (Belmont and Mitchison, 1996). The potency of stathmins suggests that they induce catastrophes through direct interactions with microtubule ends, presumably weakening the bonds of terminal subunits by inducing or stabilizing their curvature (Gupta et al., 2013).Open in a separate windowFigure 2.Proteins that recognize curved αβ-tubulin tend to make long interfaces that span both α- and β-tubulin. (A) A stathmin family protein (blue) forms a long helix that binds two αβ-tubulin heterodimers (pink and green; PDB accession no. 3RYH). (B) The structure of a complex between kinesin-1 and αβ-tubulin (PDB accession no. 4HNA) is shown with the motor in dark green and αβ-tubulin in pink and lime. Depolymerizing kinesins have insertions (red segments modeled based on a crystal structure of MCAK; PDB accession no. 1V8K), such as the KVD finger, that expand the contact region compared with purely motile kinesins. (C) The TOG1 domain (blue) from Stu2, an XMAP215 family polymerase, contacts regions of α- and β-tubulin (pink and green) that move relative to each other in the curved (left, PDB accession no. 4FFB) and straight (right, model substituting straight αβ-tubulin; PDB accession no. 1JFF) conformations of αβ-tubulin. The asterisks show where this relative movement would disrupt the TOG–tubulin interface. Red side chains indicate conserved tubulin-binding residues at the top and bottom of the TOG domain. (D) The TOG2 domain from human CLASP1 (light blue, PDB accession no. 4K92) shows an “arched” interface that in docked models like the ones shown here is not complementary to curved (left) or straight (right) conformations of αβ-tubulin. Curved and straight structures are PDB 4FFB and 1JFF, respectively. Red side chains indicate binding residues similar to those in the polymerase family TOG domains, and asterisks highlight where the arched nature of this TOG prevents a conserved binding residue from contacting its interaction partner on β-tubulin.Kinesin-13s, first identified by their central motor domain (Aizawa et al., 1992; Wordeman and Mitchison, 1995), depolymerize microtubules catalytically using the energy of ATP hydrolysis (Hunter et al., 2003). Kinesin-13s depolymerize microtubules at spindle poles to generate poleward flux (Ganem et al., 2005), at kinetochores to drive anaphase chromosome segregation (Maney et al., 1998; Rogers et al., 2004), and in neuronal processes (Homma et al., 2003). Evidence that kinesin-13s depolymerized microtubules came from the discovery of the Xenopus laevis homologue, Xkcm1, in a screen for kinesin-related proteins involved in spindle assembly (Walczak et al., 1996). Incubation of Xkcm1, also known as MCAK, with GMPCPP microtubules caused peeled protofilaments and significant “ram’s horns” structures to appear at microtubule ends (Desai et al., 1999), which indicates that MCAK binds more tightly to curved structures than to straight ones. As with all kinesins, tight binding of the motor domain is coupled to its ATP hydrolysis cycle. Kinesin-13s first bind the microtubule lattice with an on-rate constant that strongly influences its depolymerase activity (Cooper et al., 2010). Kinesin-13s then target the end of the microtubule via “lattice diffusion,” a random walk mediated by electrostatic interactions that occurs in the ADP state (Helenius et al., 2006). Exchange of ADP to ATP occurs at microtubule ends; in the ATP state, MCAK binds tightly to tubulin dimers and either induces or stabilizes their outward curvature and detachment from the microtubule lattice (Friel and Howard, 2011). The subsequent hydrolysis of ATP causes kinesin-13 to release its tubulin subunit, now detached from the lattice, and begin another cycle of depolymerization (Moores et al., 2002).A distinguishing feature of the kinesin-13 motor domain is an extension of loop L2, known as the KVD finger (Ogawa et al., 2004; Shipley et al., 2004), which protrudes from the motor domain toward the minus end of the microtubule (Fig. 2 B). Alanine substitution of the KVD motif inhibits depolymerase activity in cell-based assays (Ogawa et al., 2004) and in vitro (Shipley et al., 2004). A recent cryo-EM study showed that the kinesin-13 motor domain contacts curved tubulin on three distinct surfaces (Asenjo et al., 2013) that differ from the contact surfaces of kinesin-1 (Sindelar and Downing, 2010; Gigant et al., 2013). The location of the kinesin-13 contact surfaces could allow kinesin-13 to stabilize spontaneous curvature of tubulin dimers at either microtubule end. Alternatively, tight binding of the kinesin-13 motor domain could directly induce curvature in the tubulin dimer. In either case, by promoting curvature at the growing microtubule end, kinesin-13s weaken the association of terminal subunits and induce catastrophes.Kinesin-8s are motile depolymerases (Gupta et al., 2006; Varga et al., 2006) that establish the length of microtubules in the mitotic spindle (Goshima et al., 2005; Rizk et al., 2014), position the spindle (Gupta et al., 2006), and modulate the dynamics of kinetochore microtubules (Stumpff et al., 2008; Du et al., 2010). Unlike the nonmotile kinesin-13s, whose motor domain is fully specialized for depolymerization, kinesin-8 proteins walk to the microtubule end and remove tubulin upon arrival (Gupta et al., 2006; Varga et al., 2006). Although it is unclear if depolymerase activity is fully conserved (Du et al., 2010; Mayr et al., 2011), all kinesin-8s combine motility with a negative effect on microtubule growth. For Saccharomyces cerevisiae Kip3p, the combination of motility and depolymerase activity has a significant functional consequence: Kip3p depolymerizes longer microtubules faster than shorter ones (Varga et al., 2006). This length-dependent depolymerization can be explained by an “antenna model.” In this model, longer microtubules will accumulate more kinesin-8s, which then walk toward the microtubule end, forming length-dependent traffic jams in some cases (Leduc et al., 2012). Because the rate of depolymerization depends on the number of kinesin-8s that arrive at the microtubule end, longer microtubules will be depolymerized more quickly. The “antenna model” depends critically on the high processivity of kinesin-8, which is thought to result from an additional C-terminal microtubule-binding element (Mayr et al., 2011; Stumpff et al., 2011; Su et al., 2011; Weaver et al., 2011); the C terminus may also contribute to a recently described microtubule sliding activity in Kip3p (Su et al., 2013). Intriguingly, a single Kip3p appears to be insufficient to remove a tubulin dimer. Rather, a second Kip3p must arrive at the microtubule end to bump off the first one (Varga et al., 2009).There are less structural and mutagenesis data available to explain the unique ability of kinesin-8s to walk and depolymerize. It is also not clear that all kinesin-8s use the same cooperative mechanism described for Kip3p. Like kinesin-13, the motor domain of kinesin-8 has an extended loop L2. This loop is disordered in the available crystal structure, but has been observed to contact α-tubulin in a cryo-EM reconstruction (Peters et al., 2010). The kinesin-8 loop L2 lacks a KVD sequence, however, and systematic mutations of L2 have not yet determined its role in depolymerase activity. The extent to which kinesin-8s recognize/induce curvature at microtubule ends remains unresolved. Truncated kinesin-8 motor domains can create small peels at the ends of GMPCPP microtubules (Peters et al., 2010), which suggests that kinesin-8 can induce or stabilize curvature. The fact that two kinesin-8s are required to dissociate a tubulin subunit, however, indicates that single motors alone do not substantially weaken the bonds holding the terminal tubulin subunit. Perhaps kinesin-8s do not stabilize curved forms of αβ-tubulin as strongly as kinesin-13s do.Reconstitution of microtubule dynamics in vitro showed that the depolymerizing kinesins affect catastrophe in different ways (Gardner et al., 2011b): kinesin-13s eliminate the aging process described earlier, whereas kinesin-8s accelerate it. Importantly, the local control of catastrophes by depolymerases is accomplished primarily through the local modulation of curvature at microtubule ends.

Growth-promoting MAPs also use conformation-selective interactions with αβ-tubulin

MAPs that accelerate growth or stabilize the microtubule lattice counteract microtubule depolymerases (Tournebize et al., 2000; Kinoshita et al., 2001). XMAP215 was discovered as the major protein in Xenopus extracts that promotes microtubule growth (Gard and Kirschner, 1987). Later, functional homologues were discovered in S. cerevisiae (Stu2p) (Wang and Huffaker, 1997) and other organisms (e.g., Charrasse et al., 1998; Cullen et al., 1999). XMAP215 family proteins localize to kinetochores and microtubule organizing centers, where they contribute to chromosome movements and to spindle assembly and flux (Wang and Huffaker, 1997; Cullen et al., 1999). Loss of XMAP215 family polymerase function leads to shorter, slower-growing microtubules and often gives rise to smaller and/or aberrant spindles (Wang and Huffaker, 1997; Cullen et al., 1999). All family members contain multiple TOG domains that bind αβ-tubulin (Al-Bassam et al., 2006; Slep and Vale, 2007). The molecular mechanisms underlying the activity of these proteins, and the collective action of their arrayed TOG domains, have until recently remained obscure. Recent progress is defining the structure and biochemistry of TOG domains and their interactions with αβ-tubulin. The emerging view is that XMAP215 family polymerases, like the depolymerases, bind to curved αβ-tubulin dimers as an important part of their biochemical cycle. In this section, we will focus on the most recent developments that are shaping the molecular understanding of growth-promoting MAPs, emphasizing the somewhat better studied XMAP215 family.Affinity chromatography using immobilized TOG domains from Stu2p revealed that the TOG1 domain binds directly to unpolymerized αβ-tubulin (Al-Bassam et al., 2006). TOG domains can also bind specifically to one end of the microtubule (Al-Bassam et al., 2006). Crystal structures of TOG domains, sequence conservation, and site-directed mutagenesis defined the αβ-tubulin–interacting surface, which forms a narrow “spine” of the book-shaped domain (Al-Bassam et al., 2007; Slep and Vale, 2007).In early models for XMAP215, the arrayed TOG domains were thought to bind multiple αβ-tubulins (Gard and Kirschner, 1987). Subsequent fluorescence-based reconstitution of XMAP215 activity, however, gave results that were not consistent with this “shuttle” model (Brouhard et al., 2008). The reconstitution assays showed that XMAP215 acted processively, residing at the microtubule end long enough to perform multiple rounds of αβ-tubulin addition. Intriguingly, XMAP215 increased the rate of, but not the apparent equilibrium constant for, microtubule elongation. XMAP215 also stimulated the rate of shrinkage in the absence of unpolymerized αβ-tubulin. Similar observations were made using Alp14 (Al-Bassam et al., 2012), a Schizosaccharomyces pombe XMAP215 homologue. These studies showed that XMAP215 catalyzes polymerization: it promotes microtubule growth by using its TOG domains to repeatedly bind and stabilize an intermediate state that otherwise limits the rate of polymerization.How do TOG domains recognize the microtubule end and promote elongation? Recent structural studies (Ayaz et al., 2012, 2014) suggest that interactions with curved αβ-tubulin play a central role. The crystal structures of complexes between αβ-tubulin and the TOG1 or TOG2 domains from Stu2p revealed that both TOG domains bind to curved αβ-tubulin (Ayaz et al., 2012, 2014; Fig. 2 C). The TOG domains do not interact strongly with microtubules even though the TOG-contacting epitopes are accessible on the microtubule surface (Ayaz et al., 2012). Preferential binding to curved αβ-tubulin (Ayaz et al., 2014) occurs because the arrangement of the TOG-contacting regions of α- and β-tubulin differs between curved and straight conformations (Fig. 2 C). Conformation-selective TOG–αβ-tubulin interactions explain how XMAP215 family proteins discriminate unpolymerized αβ-tubulin from αβ-tubulin in the body of the microtubule. XMAP215 family proteins require a basic region in addition to TOG domains for microtubule plus end association and polymerase activity (Widlund et al., 2011). The polarity of TOG–αβ-tubulin interactions and the ordering of domains in the protein together explain the plus end specificity of these polymerases: only at the plus end can TOGs engage curved αβ-tubulin while the C-terminal basic region contacts surfaces deeper in the microtubule (Ayaz et al., 2012). A recent study proposed that the linked TOG domains catalyze elongation using a tethering mechanism that effectively concentrates unpolymerized αβ-tubulin near curved subunits already bound at the microtubule end (Ayaz et al., 2014). The mechanisms by which these proteins catalyze depolymerization are less understood, although depolymerization can be explained by the catalytic stabilization of an intermediate state (Brouhard et al., 2008). By analogy with the depolymerases described earlier, the stabilization of such a state by arrayed TOG domains seems likely to also depend on the preferential interactions with curved αβ-tubulin.CLASP family proteins (Pasqualone and Huffaker, 1994; Akhmanova et al., 2001) also contain TOG domains, but they are used to different effect: CLASPs do not make microtubules grow faster but instead appear to regulate the frequencies of catastrophe and rescue. For example, in vitro reconstitutions using Cls1p, a CLASP protein from S. pombe, showed that Cls1p promoted rescue (Al-Bassam et al., 2010). CLASP family proteins also localize to kinetochores and contribute to spindle flux (Maiato et al., 2005). Loss of CLASP function affects microtubule stability and causes spindle defects (Akhmanova et al., 2001; Maiato et al., 2005), but does so without significantly affecting microtubule growth rates (Mimori-Kiyosue et al., 2006). CLASPs can also stabilize microtubule bundles/overlaps (Bratman and Chang, 2007). The recently published structure of a CLASP family TOG domain (Leano et al., 2013) provided an unexpected hint about a possible origin of the different activities. Indeed, the structure revealed significant differences with XMAP215 family TOG domains even though the CLASP TOG maintains evolutionarily conserved αβ-tubulin–interacting residues (Fig. 2 D). Whereas the αβ-tubulin binding surface of XMAP215 family TOGs is relatively flat, the equivalent surface of the CLASP TOG is arched in a way that appears to break the geometric match with curved αβ-tubulin (Leano et al., 2013; Fig. 2 D). This suggests that CLASP TOG domains might bind to an even more curved conformation of αβ-tubulin that has not yet been observed, that they do not simultaneously engage α- and β-tubulin, or that they do something else. It is not yet clear how these different possibilities might contribute to the rescue-promoting activity of CLASPs. However, even though the biochemical and structural understanding of how CLASP TOGs interact with αβ-tubulin is less advanced than for XMAP215 family TOGs, the conservation of critical αβ-tubulin–interacting residues makes it seem likely that conformation-selective interactions with αβ-tubulin will play a prominent role.The modulation of microtubule dynamics by XMAP215/CLASP family proteins ensures proper microtubule function in both interphase and dividing cells. As for the depolymerases, specific interactions with curved αβ-tubulin likely underlie the different regulatory activities of XMAP215/CLASP family proteins.

Sensing conformation at lattice contacts

Thus far, we have described how microtubule polymerases and depolymerases bind selectively to curved conformations of the αβ-tubulin dimer. These interactions play a significant role in the movement of tubulin dimers into and out of the microtubule polymer. Once in the polymer, αβ-tubulin dimers make contacts with neighboring tubulins. Recently, three MAPs were shown to bind microtubules at lattice contacts: (1) the Ndc80 complex, a core kinetochore protein; (2) doublecortin (DCX), a neuronal MAP; and (3) EB1, the canonical end-binding protein. Here we will summarize recent progress demonstrating how these proteins recognize distinctive features of lattice contacts.The Ndc80 complex is a core component of the kinetochore–microtubule interface (Janke et al., 2001; Wigge and Kilmartin, 2001; McCleland et al., 2003), forming a “sleeve” that connects the outer kinetochore to microtubules of the mitotic spindle (Cheeseman et al., 2006; DeLuca et al., 2006). Loss of Ndc80 function leads to chromosome segregation errors in mitosis (McCleland et al., 2004; DeLuca et al., 2005). Ndc80 binds to microtubules at the longitudinal interface between α- and β-tubulin and extends outward toward the plus end at an ∼60° angle (Cheeseman et al., 2006; Wilson-Kubalek et al., 2008). Ndc80 binds to both the intradimer and interdimer interface and forms oligomeric arrays (Alushin et al., 2010). The binding of Ndc80 to this longitudinal lattice contact may confer a preference for straight rather than curved microtubule lattices, because the shape of the Ndc80 binding site is expected to change as a protofilament bends (Alushin et al., 2010; Fig. 3 A). Preferential binding to straight protofilaments might allow the Ndc80 complex to remain attached to the end of a shrinking microtubule. Indeed, reconstitutions of the Ndc80 complex interacting with dynamic microtubules show that the curved shrinking end acts as a “reflecting wall,” giving rise to “biased diffusion” (Powers et al., 2009). Interestingly, the Ndc80 complex also promotes rescue (Umbreit et al., 2012), and selective binding to straight lattice contacts may contribute to this rescue activity.Open in a separate windowFigure 3.Proteins that bind microtubules can distinguish unique configurations at lattice contacts. (A) Ndc80 (light and dark blue) binds the contact within (dark blue) and between (light blue) αβ-tubulin heterodimers (pink and green). The left shows part of an Ndc80 array on straight protofilaments (PDB accession no. 3IZ0). The right shows that neighboring Ndc80 molecules clash when modeled onto a curved protofilament. Individual Ndc80s may read the conformation at a single joint, or the change in conformation may disrupt cooperative interactions between adjacent Ndc80s. (B) Two views of DCX (blue) binding a lattice contact at the vertex of four αβ-tubulins, PDB accession no. 4ATU. Cooperative interactions on the microtubule allow DCX to discriminate between the subtle changes that accompany different protofilament numbers (11: orange, EMDataBank [EMD] accession no. 5191; 13: red, EMD accession no. 5193; 15: yellow, EMD accession no. 5195). (C) EB1 (left, dark blue) binds at the same vertex as DCX (PDB accession no. 4AB0), but EB1 binds preferentially to GTP vertices over GDP vertices, and is not sensitive to protofilament number. The same section of microtubule with EB1 removed (right) shows the location of nucleotide-dependent changes at the four-way vertex: helix H3 of β-tubulin (red patch at the lower right of the four-way junction), and the intermediate (Int.) domain of α-tubulin (yellow patch at the top left of the four-way junction). pfs, protofilaments.DCX, a MAP expressed in developing neurons (Francis et al., 1999; Gleeson et al., 1999) and mutated in cases of subcortical band heterotopia (des Portes et al., 1998; Gleeson et al., 1998), is unique in its ability to bind specifically to 13-protofilament microtubules over other protofilament numbers (Moores et al., 2004; Fig. 3 B). DCX contains two nonidentical, microtubule-binding “DC” domains (Taylor et al., 2000) that share a ubiquitin-like fold (Kim et al., 2003). A cryo-EM reconstruction showed that a single DC domain binds to microtubules at the vertex of four tubulin dimers in the so-called “B” lattice configuration (Fourniol et al., 2010). The DCX binding site is ideally situated to detect the subtle changes at lattice contacts that result from different protofilament numbers, which range from 11 to 16 for mammalian microtubules (Sui and Downing, 2010). Despite their ideal location, protofilament preference is not a property of single DCX molecules. Rather, it is cooperative interactions between neighboring DCX molecules that are sensitive to the spacing between protofilaments (Bechstedt and Brouhard, 2012). In vitro, this selectivity enables DCX to nucleate homogeneous, 13-protofilament microtubules (Moores et al., 2004). The function of DCX in developing neurons remains unclear, with models ranging from microtubule stabilization (Gleeson et al., 1999) to regulation of kinesin traffic (Liu et al., 2012).EB1, the canonical end-binding protein (Morrison et al., 1998), uses its calponin homology (CH) domain (Hayashi and Ikura, 2003) to bind the same lattice contact as DCX (Maurer et al., 2012). EB1 forms “comets” by binding rapidly and tightly to a distinct feature at the growing microtubule end but only weakly to the “mature” lattice (Bieling et al., 2007). Recent work has defined this distinctive feature as the nucleotide state. EB1 binds preferentially to microtubules built from GTP analogues (Zanic et al., 2009; Maurer et al., 2011). Combined with careful analysis of the size, shape, and dynamics of EB1 comets (Bieling et al., 2007), these results established that EB1 recognizes microtubule ends by binding specifically to the “GTP cap,” which is an extended region of the microtubule end that is enriched with GTP- and GDP-Pi-tubulin dimers. A recent cryo-EM reconstruction of the CH domain of Mal3 (the S. pombe EB1) bound to GTPγS microtubules provided a possible structural mechanism for how EB1 might differentiate GTP from GDP lattices (Maurer et al., 2012; Fig. 3 C). Mal3 was observed to contact helix H3 of β-tubulin, which connects directly to the exchangeable nucleotide-binding site. EB1 also contacts the regions of α-tubulin that move during the compaction of the lattice that follows GTP hydrolysis (Alushin et al., 2014). Mutation of conserved EB1 residues that contact either helix H3 or the compacting region of α-tubulin disrupts the end-tracking behavior of EB1 (Slep and Vale, 2007; Maurer et al., 2012). Interactions with helix H3 and the compacting region of α-tubulin also enable EB1 to accelerate the transitions of tubulin from the GTP state to the GDP state; in other words, EB1 acts as a “maturation factor” for the microtubule end (Maurer et al., 2014). EB1 recruits a large network of plus-end-tracking proteins (Akhmanova and Steinmetz, 2008) through interactions with the EB1 C terminus (Hayashi et al., 2005; Honnappa et al., 2006) and EB1 homology domain (Honnappa et al., 2009). This diverse and complex protein network is essential for the regulation of microtubule dynamics, the capture of microtubule ends by the cell cortex (Kodama et al., 2003) and endoplasmic reticulum (Grigoriev et al., 2008), and the positioning of the mitotic spindle (Liakopoulos et al., 2003).As mentioned earlier, microtubule ends also show unique structural configurations, namely tapered, outwardly flared, and flattened structures collectively described as “sheets” (Chrétien et al., 1995). The sheets contain distinctive lattice contacts, and recent work shows that the microtubule-binding activities of DCX and EB1 are sensitive to these structural features. DCX, for example, binds specifically to the outwardly flared sheets (Bechstedt et al., 2014), which enables DCX to track microtubule ends. Evidence for the ability of EB1 to recognize or control a distinct lattice configuration comes from the reconstitutions showing that EB1 promotes elongation synergistically with XMAP215 (Zanic et al., 2013): lack of a detectable direct EB1–XMAP215 interaction suggested that the observed synergy was mediated through alterations of the microtubule end structure itself. Further evidence that EB1 can affect the structure of the microtubule lattice comes from data showing that EB1 can nucleate “A” lattice microtubules in vitro (des Georges et al., 2008) and influence protofilament number distributions (Vitre et al., 2008; Maurer et al., 2012). The connection between the structure of microtubule ends, their nucleotide state, and microtubule dynamics is an important open question.

Conclusions and outlook

The αβ-tubulin dimer adopts a range of conformations as it moves in and out of the microtubule polymer, including changes to its intrinsic curvature and changes to its lattice contacts. These different conformations affect microtubule dynamics by altering the strength of lattice association and the rate of GTP hydrolysis. The work we discussed here has revealed an intimate linkage between these different conformations and the activities of key proteins that regulate microtubule dynamics. It is now clear that selective interactions with distinct conformations of unpolymerized and polymerized αβ-tubulin define the cell physiology of the microtubule cytoskeleton. Recently developed methods for purifying or overexpressing αβ-tubulin (des Georges et al., 2008; Johnson et al., 2011; Widlund et al., 2012; Minoura et al., 2013) are facilitating structural studies and allowing the biochemistry of αβ-tubulin polymerization to be dissected in unprecedented detail. Microtubule structural biology is entering a golden age, where the pace of new structural information is accelerating. We anticipate that future crystallographic and high-resolution cryo-EM studies will define the strategies used by other MAPs to recognize and control the conformation of αβ-tubulin, and may reveal new conformations of αβ-tubulin inside and outside of the microtubule. Reconstitutions of microtubule dynamics are rapidly increasing in complexity and are beginning to reveal how the activities of multiple MAPs can reinforce or antagonize each other (Zanic et al., 2013). More complex reconstitutions are also defining the minimal requirements for creating cellular-scale structures like the mitotic spindle (Bieling et al., 2010; Subramanian et al., 2013). Reconstitutions will also greatly advance the understanding of the dynamics and regulation of microtubule minus ends. As the ever-advancing structural data are integrated with reconstitution data, incorporated into computational models, and correlated with cell biology experiments, a robust, multiscale understanding of microtubule biology will come within reach.  相似文献   

15.
Genetic Mechanisms of Coffee Extract Protection in a Caenorhabditis elegans Model of β-Amyloid Peptide Toxicity     
Vishantie Dostal  Christine M. Roberts  Christopher D. Link 《Genetics》2010,186(3):857-866
  相似文献   

16.
Immunomodulation with Recombinant Interferon-γ1b in Pulmonary Tuberculosis     
Rod Dawson  Rany Condos  Doris Tse  Maryann L. Huie  Stanley Ress  Chi-Hong Tseng  Clint Brauns  Michael Weiden  Yoshihiko Hoshino  Eric Bateman  William N. Rom 《PloS one》2009,4(9)

Background

Current treatment regimens for pulmonary tuberculosis require at least 6 months of therapy. Immune adjuvant therapy with recombinant interferon-γ1b (rIFN-γb) may reduce pulmonary inflammation and reduce the period of infectivity by promoting earlier sputum clearance.

Methodology/Principal Findings

We performed a randomized, controlled clinical trial of directly observed therapy (DOTS) versus DOTS supplemented with nebulized or subcutaneously administered rIFN-γ1b over 4 months to 89 patients with cavitary pulmonary tuberculosis. Bronchoalveolar lavage (BAL) and blood were sampled at 0 and 4 months. There was a significant decline in levels of inflammatory cytokines IL-1β, IL-6, IL-8, and IL-10 in 24-hour BAL supernatants only in the nebulized rIFN-γ1b group from baseline to week 16. Both rIFN-γ1b groups showed significant 3-fold increases in CD4+ lymphocyte response to PPD at 4 weeks. There was a significant (p = 0.03) difference in the rate of clearance of Mtb from the sputum smear at 4 weeks for the nebulized rIFN-γ1b adjuvant group compared to DOTS or DOTS with subcutaneous rIFN-γ1b. In addition, there was significant reduction in the prevalence of fever, wheeze, and night sweats at 4 weeks among patients receiving rFN-γ1b versus DOTS alone.

Conclusion

Recombinant interferon-γ1b adjuvant therapy plus DOTS in cavitary pulmonary tuberculosis can reduce inflammatory cytokines at the site of disease, improve clearance of Mtb from the sputum, and improve constitutional symptoms.

Trial Registration

ClinicalTrials.gov NCT00201123  相似文献   

17.
Notes: β(1-3)Glucanosyltransferase Gel4p Is Essential for Aspergillus fumigatus     
Amandine Gastebois  Thierry Fontaine  Jean-Paul Latgé  Isabelle Mouyna 《Eukaryotic cell》2010,9(8):1294-1298
The β(1-3)glucanosyltransferase GEL family of Aspergillus fumigatus contains 7 genes, among which only 3 are expressed during mycelial growth. The role of the GEL4 gene was investigated in this study. Like the other Gelps, it encodes a glycosylphosphatidylinositol (GPI)-anchored protein. In contrast to the other β(1-3)glucanosyltransferases analyzed to date, it is essential for this fungal species.β(1-3)Glucan is the main component of the fungal cell wall (11). In fungi, β(1-3)glucans are synthesized by a plasma membrane-bound glucan synthase complex. Neosynthesized glucans are then extruded into the periplasmic space (2, 3, 9), where they become branched and covalently linked to other cell wall components, resulting in the formation of three-dimensional rigid structures. In the search of transglycosidase in the filamentous fungus Aspergillus fumigatus, β(1-3)glucanosyltransferases were identified and classified as a unique family (GH72) in the Carbohydrate-Active enZYmes database (http://www.cazy.org/). These enzymes cleave the β(1-3) bond of a β(1-3)glucan oligosaccharide with at least 10 glucose units and transfer the newly formed reducing end (>5 glucose units) to the nonreducing end of another β(1-3)glucan oligosaccharide, resulting in the elongation of the β(1-3)glucans. This reaction can proceed in vitro until the neosynthetized β(1-3)glucan becomes insoluble. Initially demonstrated biochemically, the requirement for long-chain β(1-3)glucan oligosaccharide has now been confirmed by the analysis of the first crystal structure obtained in this transglycosidase family (7, 8). First discovered in Aspergillus fumigatus and named Gelp for glucan elongase, this activity has been found in all fungal species investigated to date and could be assigned to orthologous proteins, such as Gasp or Phrp, that were known to be involved in cell wall integrity but were endowed with an unknown biochemical function (12, 13, 14).  相似文献   

18.
Specific α- and β-Tubulin Isotypes Optimize the Functions of Sensory Cilia in Caenorhabditis elegans     
Daryl D. Hurd  Renee M. Miller  Lizbeth Nú?ez  Douglas S. Portman 《Genetics》2010,185(3):883-896
Primary cilia have essential roles in transducing signals in eukaryotes. At their core is the ciliary axoneme, a microtubule-based structure that defines cilium morphology and provides a substrate for intraflagellar transport. However, the extent to which axonemal microtubules are specialized for sensory cilium function is unknown. In the nematode Caenorhabditis elegans, primary cilia are present at the dendritic ends of most sensory neurons, where they provide a specialized environment for the transduction of particular stimuli. Here, we find that three tubulin isotypes—the α-tubulins TBA-6 and TBA-9 and the β-tubulin TBB-4—are specifically expressed in overlapping sets of C. elegans sensory neurons and localize to the sensory cilia of these cells. Although cilia still form in mutants lacking tba-6, tba-9, and tbb-4, ciliary function is often compromised: these mutants exhibit a variety of sensory deficits as well as the mislocalization of signaling components. In at least one case, that of the CEM cephalic sensory neurons, cilium architecture is disrupted in mutants lacking specific ciliary tubulins. While there is likely to be some functional redundancy among C. elegans tubulin genes, our results indicate that specific tubulins optimize the functional properties of C. elegans sensory cilia.THE fitness of all organisms depends on an ability to appropriately sense and respond to the environment. At the cellular level, many specific architectures have evolved to optimize these sensory functions. Prominent among these is the sensory cilium, a tubulin-based cytoplasmic extension that interrogates the extracellular environment in many biological contexts (Davenport and Yoder 2005; Berbari et al. 2009). Cilia are important for the transduction of a broad range of visual, auditory, mechanical, thermal, and chemical stimuli. They also function during development to receive a variety of signals, both chemical and mechanical, that regulate proliferation and differentiation (Goetz and Anderson 2010). Indeed, the disruption of cilium assembly and function can give rise to a spectrum of human diseases collectively known as ciliopathies (Berbari et al. 2009; Lancaster and Gleeson 2009). These disorders, which include autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), Bardet–Biedl syndrome, Meckel–Gruber syndrome, and Joubert syndrome, are associated with a variety of pathogenic conditions including polycystic kidneys and neurological impairments.At the core of all cilia and flagella is the microtubule axoneme. This characteristic structural element comprises nine doublet outer microtubules that may surround a central pair, the presence of which often indicates a motile cilium/flagellum. Like all microtubule-based structures, ciliary axonemes are built of heterodimers of α- and β-tubulins, highly conserved small GTP-binding proteins. The recruitment of other cilium components, including signal transduction machinery, requires a conserved assembly and maintenance process called intraflagellar transport (IFT) (Blacque et al. 2008; Pedersen and Rosenbaum 2008). IFT employs two major complexes that transport ciliary cargo bidirectionally by traveling along the axonemal microtubules. Loss of individual IFT components can cause a broad spectrum of defects in the assembly, maintenance, and function of cilia.Important insights into cilium structure and function have come from studies of genetically tractable organisms, particularly the green alga Chlamydomonas and the nematode Caenorhabditis elegans (Bae and Barr 2008; Pedersen and Rosenbaum 2008). In C. elegans, sensory cilia are found exclusively at the dendritic ends of sensory neurons. These cilia constitute a highly specialized sensory environment characterized by localized sensory receptors and specific signaling components. Cilium morphology is quite distinctive in many of these cells and likely contributes to their functional specialization (Ward et al. 1975). Recent progress has shed light on the mechanisms that confer this specialization onto more general pan-ciliary pathways (Evans et al. 2006; Mukhopadhyay et al. 2007; Jauregui et al. 2008; Mukhopadhyay et al. 2008; Silverman and Leroux 2009).The genomes of many eukaryotes harbor multiple α- and β-tubulin genes. Two hypotheses, which are not mutually exclusive, have been proposed to account for these paralogs (Cleveland 1987; Wade 2007). At one extreme, different tubulin isotypes might be functionally redundant, such that their minor coding differences are largely irrelevant. According to this model, multiple genes allow the maintenance of a stable pool of available monomers and dimers. The small amount of sequence variation within the α- and β-tubulin families supports this idea, as do studies of functionally redundant mitotic tubulins in C. elegans (Ellis et al. 2004; Lu et al. 2004; Phillips et al. 2004; Lu and Mains 2005). The alternative hypothesis proposes that specific structures, e.g., ciliary axonemes or axonal microtubules, rely on tubulins optimized for specific roles. Support for this idea has come from studies of cultured mammalian neurons (Joshi and Cleveland 1989), Drosophila (Hutchens et al. 1997; Raff et al. 1997), and human tubulins (Vent et al. 2005; Jaglin et al. 2009). In Drosophila, studies of motile sperm flagella have revealed that the sperm-specific β2 tubulin isoform builds not only the specialized motile axoneme but also all other tubulin-based structures (Kemphues et al. 1982). However, sequences both within and outside the axoneme motif in the C-terminal tail of this tubulin isoform are required for the flagellar axoneme, and other closely related β-tubulins cannot support this role (Fuller et al. 1987; Raff et al. 1997; Popodi et al. 2008). Genetic interactions have provided evidence that β2 tubulin heterodimerizes with the α-tubulin 84B (Hays et al. 1989), which also possesses specific functional properties not provided by structurally similar α-tubulins (Hutchens et al. 1997). In C. elegans, a specific role for tubulin isoforms has been described in the six touch receptor neurons. These nonciliated cells harbor unusual 15-filament microtubules composed of dimers of the α-tubulin MEC-12 and the β-tubulin MEC-7. The loss of mec-7 or mec-12, the expression of which is largely restricted to these cells, results in the conversion of 15-filament microtubules to the standard 11-microfilament variety and a commensurate loss of light-touch response (Savage et al. 1989; Fukushige et al. 1999; Bounoutas et al. 2009). Thus experimental support exists for both of these opposing views, and it seems likely that the role of specific tubulin isoforms in regulating microtubule structure and function differs according to cell and organelle type.The C. elegans genome encodes nine α- and six β-tubulin genes (Gogonea et al. 1999). Some of these genes, particularly tba-1, tba-2, tbb-1, and tbb-2, are expressed broadly during embryogenesis and function redundantly in spindle assembly and positioning (Ellis et al. 2004; Lu et al. 2004; Phillips et al. 2004; Lu and Mains 2005). tba-1 and tbb-2 have also been recently shown to be important for axon outgrowth and synaptogenesis (Baran et al. 2010). Several others, including mec-7, mec-12, and the β-tubulin ben-1, have been identified through genetic screens for particular phenotypes, such as touch insensitivity or benzimidazole resistance (Driscoll et al. 1989; Savage et al. 1989; Fukushige et al. 1999). However, the extent to which specific tubulin isoforms are required for structural and functional diversity in the C. elegans nervous system remains unknown. Here, taking advantage of several existing genome-wide data sets, we identify the α-tubulins TBA-6 and TBA-9 and the β-tubulin TBB-4 as strong candidates for tubulins that have roles in sensory cilia. We find that each of these genes are expressed in characteristic, partially overlapping, sets of sensory neurons, where their products localize to ciliary axonemes. While the loss of any one (or all three) of these genes does not abolish ciliogenesis, tubulin mutants exhibit significant defects in the localization of cilium proteins and in some cilium-dependent behavioral responses. Together, our results indicate that specific α- and β-tubulin isoforms are important, although not essential, for the efficient assembly and function of specific classes of C. elegans sensory cilia. Sensory cilia throughout the animal kingdom may therefore employ specific tubulin isoforms to optimize their function.  相似文献   

19.
Genetic Variants of the α-Synuclein Gene SNCA Are Associated with Multiple System Atrophy     
Ammar Al-Chalabi  Alexandra Dürr  Nicholas W. Wood  Michael H. Parkinson  Agnes Camuzat  Jean-Sébastien Hulot  Karen E. Morrison  Alan Renton  Sigurd D. Sussmuth  Bernhard G. Landwehrmeyer  Albert Ludolph  Yves Agid  Alexis Brice  P. Nigel Leigh  Gilbert Bensimon  for the NNIPPS Genetic Study Group 《PloS one》2009,4(9)

Background

Multiple system atrophy (MSA) is a progressive neurodegenerative disorder characterized by parkinsonism, cerebellar ataxia and autonomic dysfunction. Pathogenic mechanisms remain obscure but the neuropathological hallmark is the presence of α-synuclein-immunoreactive glial cytoplasmic inclusions. Genetic variants of the α-synuclein gene, SNCA, are thus strong candidates for genetic association with MSA. One follow-up to a genome-wide association of Parkinson''s disease has identified association of a SNP in SNCA with MSA.

Methodology/Findings

We evaluated 32 SNPs in the SNCA gene in a European population of 239 cases and 617 controls recruited as part of the Neuroprotection and Natural History in Parkinson Plus Syndromes (NNIPPS) study. We used 161 independently collected samples for replication. Two SNCA SNPs showed association with MSA: rs3822086 (P = 0.0044), and rs3775444 (P = 0.012), although only the first survived correction for multiple testing. In the MSA-C subgroup the association strengthened despite more than halving the number of cases: rs3822086 P = 0.0024, OR 2.153, (95% CI 1.3–3.6); rs3775444 P = 0.0017, OR 4.386 (95% CI 1.6–11.7). A 7-SNP haplotype incorporating three SNPs either side of rs3822086 strengthened the association with MSA-C further (best haplotype, P = 8.7×10−4). The association with rs3822086 was replicated in the independent samples (P = 0.035).

Conclusions/Significance

We report a genetic association between MSA and α-synuclein which has replicated in independent samples. The strongest association is with the cerebellar subtype of MSA.

Trial Registration

ClinicalTrials.gov NCT00211224. [NCT00211224]  相似文献   

20.
Elevated Genome-Wide Instability in Yeast Mutants Lacking RNase H Activity     
Karen O’Connell  Sue Jinks-Robertson  Thomas D. Petes 《Genetics》2015,201(3):963-975
  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