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 523 毫秒
1.
Caffeic acid O-methyltransferase (COMT) is a bifunctional enzyme that methylates the 5- and 3-hydroxyl positions on the aromatic ring of monolignol precursors, with a preference for 5-hydroxyconiferaldehyde, on the way to producing sinapyl alcohol. Lignins in COMT-deficient plants contain benzodioxane substructures due to the incorporation of 5-hydroxyconiferyl alcohol (5-OH-CA), as a monomer, into the lignin polymer. The derivatization followed by reductive cleavage method can be used to detect and determine benzodioxane structures because of their total survival under this degradation method. Moreover, partial sequencing information for 5-OH-CA incorporation into lignin can be derived from detection or isolation and structural analysis of the resulting benzodioxane products. Results from a modified derivatization followed by reductive cleavage analysis of COMT-deficient lignins provide evidence that 5-OH-CA cross couples (at its β-position) with syringyl and guaiacyl units (at their O-4-positions) in the growing lignin polymer and then either coniferyl or sinapyl alcohol, or another 5-hydroxyconiferyl monomer, adds to the resulting 5-hydroxyguaiacyl terminus, producing the benzodioxane. This new terminus may also become etherified by coupling with further monolignols, incorporating the 5-OH-CA integrally into the lignin structure.Lignins are polymeric aromatic constituents of plant cell walls, constituting about 15% to 35% of the dry mass (Freudenberg and Neish, 1968; Adler, 1977). Unlike other natural polymers such as cellulose or proteins, which have labile linkages (glycosides and peptides) between their building units, lignins’ building units are combinatorially linked with strong ether and carbon-carbon bonds (Sarkanen and Ludwig, 1971; Harkin, 1973). It is difficult to completely degrade lignins. Lignins are traditionally considered to be dehydrogenative polymers derived from three monolignols, p-coumaryl alcohol 1h (which is typically minor), coniferyl alcohol 1g, and sinapyl alcohol 1s (Fig. 1; Sarkanen, 1971). They can vary greatly in their composition in terms of their plant and tissue origins (Campbell and Sederoff, 1996). This variability is probably determined and regulated by different activities and substrate specificities of the monolignol biosynthetic enzymes from different sources, and by the carefully controlled supply of monomers to the lignifying zone (Sederoff and Chang, 1991).Open in a separate windowFigure 1.The monolignols 1, and marker compounds 2 to 4 resulting from incorporation of novel monomer 15h into lignins: thioacidolysis monomeric marker 2, dimers 3, and DFRC dimeric markers 4.Recently there has been considerable interest in genetic modification of lignins with the goal of improving the utilization of lignocellulosics in various agricultural and industrial processes (Baucher et al., 2003; Boerjan et al., 2003a, 2003b). Studies on mutant and transgenic plants with altered monolignol biosynthesis have suggested that plants have a high level of metabolic plasticity in the formation of their lignins (Sederoff et al., 1999; Ralph et al., 2004). Lignins in angiosperm plants with depressed caffeic acid O-methyltransferase (COMT) were found to derive from significant amounts of 5-hydroxyconiferyl alcohol (5-OH-CA) monomers 15h (Fig. 1) substituting for the traditional monomer, sinapyl alcohol 1s (Marita et al., 2001; Ralph et al., 2001a, 2001b; Jouanin et al., 2004; Morreel et al., 2004b). NMR analysis of a ligqnin from COMT-deficient poplar (Populus spp.) has revealed that novel benzodioxane structures are formed through β-O-4 coupling of a monolignol with 5-hydroxyguaiacyl units (resulting from coupling of 5-OH-CA), followed by internal trapping of the resultant quinone methide by the phenolic 5-hydroxyl (Ralph et al., 2001a). When the lignin was subjected to thioacidolysis, a novel 5-hydroxyguaiacyl monomer 2 (Fig. 1) was found in addition to the normal guaiacyl and syringyl thioacidolysis monomers (Jouanin et al., 2000). Also, a new compound 3g (Fig. 1) was found in the dimeric products from thioacidolysis followed by Raney nickel desulfurization (Lapierre et al., 2001; Goujon et al., 2003).Further study with the lignin using the derivatization followed by reductive cleavage (DFRC) method also confirmed the existence of benzodioxane structures, with compounds 4 (Fig. 1) being identified following synthesis of the authentic parent compounds 9 (Fig. 2). However, no 5-hydroxyguaiacyl monomer could be detected in the DFRC products. These facts imply that the DFRC method leaves the benzodioxane structures fully intact, suggesting that the method might therefore be useful as an analytical tool for determining benzodioxane structures that are linked by β-O-4 ethers. Using a modified DFRC procedure, we report here on results that provide further evidence for the existence of benzodioxane structures in lignins from COMT-deficient plants, that 5-OH-CA is behaving as a rather ideal monolignol that can be integrated into plant lignins, and demonstrate the usefulness of the DFRC method for determining these benzodioxane structures.Open in a separate windowFigure 2.Synthesis of benzodioxane DFRC products 12 (see later in Fig. 6 for their structures). i, NaH, THF. ii, Pyrrolidine. iii, 1g or 1s, benzene/acetone (4/1, v/v). iv, DIBAL-H, toluene. v, Iodomethane-K2CO3, acetone. vi, Ac2O pyridine.  相似文献   

2.
Dynein is a microtubule-based molecular motor that is involved in various biological functions, such as axonal transport, mitosis, and cilia/flagella movement. Although dynein was discovered 50 years ago, the progress of dynein research has been slow due to its large size and flexible structure. Recent progress in understanding the force-generating mechanism of dynein using x-ray crystallography, cryo-electron microscopy, and single molecule studies has provided key insight into the structure and mechanism of action of this complex motor protein.It has been 50 years since dynein was discovered and named by Ian Gibbons as a motor protein that drives cilia/flagella bending (Gibbons, 1963; Gibbons and Rowe, 1965). In the mid-1980s, dynein was also found to power retrograde transport in neurons (Paschal and Vallee, 1987). Subsequently, the primary amino acid sequence of the cytoplasmic dynein heavy chain, which contains the motor domain, was determined from the cDNA sequence (Mikami et al., 1993; Zhang et al., 1993). Like other biological motors, such as kinesins and myosins, the amino acid sequence of the dynein motor domain is well conserved. There are 16 putative genes that encode dynein heavy chains in the human genome (Yagi, 2009). Among these is one gene encoding cytoplasmic dynein heavy chain and one encoding retrograde intraflagellar transport dynein heavy chain, while the rest encode for heavy chains of axonemal dyneins. Most of the genes encoding the human dynein heavy chain have a counterpart in Chlamydomonas reinhardtii, which suggests that their functions are conserved from algae to humans.Dynein is unique compared with kinesin and myosin because dynein molecules form large molecular complexes. For example, one axonemal outer arm dynein molecule of C. reinhardtii is composed of three dynein heavy chains, two intermediate chains, and more than ten light chains (King, 2012). Mammalian cytoplasmic dynein consists of two heavy chains and several smaller subunits (Fig. 1 A; Vallee et al., 1988; Allan, 2011). The cargoes of cytoplasmic dynein are various membranous organelles, including lysosomes, endosomes, phagosomes, and the Golgi complex (Hirokawa, 1998). It is likely that one cytoplasmic dynein heavy chain can adapt to diverse cargos and functions by changing its composition.Open in a separate windowFigure 1.Atomic structures of cytoplasmic dynein. (A) Schematic structure of cytoplasmic dynein complex, adapted from Allan (2011). (B) The primary structure of cytoplasmic dynein. (C and D) The atomic model of D. discoideum cytoplasmic dynein motor domain (PDB accession no. 3VKG) overlaid on a microtubule (EMDB-5193; Sui and Downing, 2010) according to the orientation determined by Mizuno et al. (2007) (C) Side view. (D) View from the plus end of microtubule. (E) Schematic domain structure of dynein.Dynein must have a distinct motor mechanism from kinesin and myosin, because it belongs to the AAA+ family of proteins and does not have the conserved amino acid motifs, called the switch regions, present in kinesins, myosins, and guanine nucleotide-binding proteins (Vale, 1996). Therefore, studying dynein is of great interest because it will reveal new design principles of motor proteins. This review will focus on the mechanism of force generation by cytoplasmic and axonemal dynein heavy chains revealed by recent structural and biophysical studies.

Anatomy of dynein

To understand the chemomechanical cycle of dynein based on its molecular structure, it is important to obtain well-diffracting crystals and build accurate atomic models. Recently, Kon and colleagues determined the crystal structures of Dictyostelium discoideum cytoplasmic dynein motor domain, first at 4.5-Å resolution (Kon et al., 2011), and subsequently at 2.8 Å (without the microtubule binding domain) and 3.8-Å (wild type) resolution (Kon et al., 2012). Carter and colleagues also determined the crystal structures of the Saccharomyces cerevisiae (yeast) cytoplasmic dynein motor domain, first at 6-Å resolution (Carter et al., 2011), and later at 3.3–3.7-Å resolution (Schmidt et al., 2012). According to these crystal structures as well as previous EM studies, the overall structure of the dynein heavy chain is divided into four domains: tail, linker, head, and stalk (Fig. 1, B–E). Simply put, each domain carries out one essential function of a motor protein: the tail is the cargo binding domain, the head is the site of ATP hydrolysis, the linker is the mechanical amplifier, and the stalk is the track-binding domain.The tail, which is not part of the motor domain and is absent from crystal structures, is located at the N-terminal ∼1,400 amino acid residues and involved in cargo binding (gray in Fig. 1, B and E). The next ∼550 residues comprise the “linker” (pink in Fig. 1, B–E), which changes its conformation depending on the nucleotide state (Burgess et al., 2003; Kon et al., 2005). This linker domain was first observed by negative staining EM in combination with single particle analysis of dynein c, an isoform of inner arm dynein from C. reinhardtii flagella (Burgess et al., 2003). According to the crystal structures, the linker is made of bundles of α-helices and lies across the AAA+ head domain, forming a 10-nm-long rod-like structure (Fig. 1, C and D). Recent class averaged images of D. discoideum cytoplasmic dynein show that the linker domain is stiff along its entire length when undocked from the head (Roberts et al., 2012). The head (motor) domain of dynein is composed of six AAA+ (ATPase associated with diverse cellular activities) modules (Neuwald et al., 1999; color-coded in Fig. 1, B–E). Although many AAA+ family proteins are a symmetric homohexamer (Ammelburg et al., 2006), the AAA+ domains of dynein are encoded by a single heavy chain gene and form an asymmetric heterohexamer. Among the six AAA+ domains, hydrolysis at the first AAA domain mainly provides the energy for dynein motility (Imamula et al., 2007; Kon et al., 2012). The hexameric ring has two distinct faces: the linker face and the C-terminal face. The linker face is slightly convex and the linker domain lies across this side (Fig. 1 D, left side). The other side of the ring has the C-terminal domain (Fig. 1 D, right side).The stalk domain of dynein was identified as the microtubule-binding domain (MTBD; Gee et al., 1997). It emanates from the C-terminal face of AAA4 and is composed of antiparallel α-helical coiled-coil domain (yellow in Fig. 1, B–E). The tip of the stalk is the actual MTBD. Interestingly, the crystal structures revealed another antiparallel α-helical coiled coil that emerges from AAA5 (orange in Fig. 1, B–E), and this region is called the buttress (Carter et al., 2011) or strut (Kon et al., 2011), which was also observed as the bifurcation of the stalk by negative-staining EM (Burgess et al., 2003; Roberts et al., 2009). The tip of the buttress/strut is in contact with the middle of the stalk and probably works as a mechanical reinforcement of the stalk.

The chemomechanical cycle of dynein

Based on structural and biochemical data, a putative chemomechanical cycle of dynein is outlined in Fig. 2 (A–E). In the no-nucleotide state, dynein is bound to a microtubule through its stalk domain, and its tail region is bound to cargoes (Fig. 2 A). The crystal structures of yeast dynein are considered to be in this no-nucleotide state. When ATP is bound to the AAA+ head, the MTBD quickly detaches from the microtubule (Fig. 2 B; Porter and Johnson, 1983). The ATP binding also induces “hinging” of the linker from the head (Fig. 2 C). According to the biochemical analysis of recombinant D. discoideum dynein (Imamula et al., 2007), the detachment from the microtubule (Fig. 2, A and B) is faster than the later hinging (Fig. 2, B and C). As a result of these two reactions, the head rotates or shifts toward the minus end of the microtubule (for more discussion about “rotate” versus “shift” see the “Dyneins in the axoneme” section) and the MTBD steps forward. The directionality of stepping seems to be mainly determined by the MTBD, because the direction of dynein movement does not change even if the head domain is rotated relative to the microtubule by insertion or deletion of the stalk (Carter et al., 2008). In the presence of ADP and vanadate, dynein is considered to be in this state (Fig. 2 C).Open in a separate windowFigure 2.Presumed chemomechanical cycle and stepping of dynein. (A–E) Chemomechanical cycle of dynein. The pre- and post-power stroke states are also called the primed and unprimed states, respectively. The registries of the stalk coiled coil are denoted as α and β according to Gibbons et al. (2005). (F and G) Processive movement of kinesin (F) and dynein (G). (F) Hand-over-hand movement of kinesin. A step by one head (red) is always followed by the step of another head (green). The stepping of kinesin is on one protofilament of microtubule. (G) Presumed stepping of dynein. The step size varies and the interhead separation can be large. A step by one head (red) is not always flowed by the step of another head (green). (H) A model of strain-based dynein ATPase activation. (G, top) Without strain, the gap between the AAA1 and AAA2 is open and the motor domain cannot hydrolyze ATP. (G, bottom) Under a strain imposed between MTBD and tail (thin black arrows), the gap becomes smaller (thick black arrows) and turns on ATP hydrolysis by dynein.After the MTBD rebinds to the microtubule at the forward site (Fig. 2 D), release of hydrolysis products from the AAA+ head is activated (Holzbaur and Johnson, 1989) and the hinged linker goes back to the straight conformation (Fig. 2 E; Kon et al., 2005). The crystal structure of D. discoideum dynein is considered to be in the state after phosphate release and before ADP release. This straightening of the linker is considered to be the power-generating step and brings the cargo forward relative to the microtubule.

The MTBD of dynein

As outlined in Fig. 2, the nucleotide state of the head domain may control the affinity of the MTBD to the microtubule. Conversely, the binding of the MTBD to the microtubule should activate the ATPase activity of the head domain. This two-way communication is transmitted through the simple ∼17-nm-long α-helical coiled-coil stalk and the buttress/strut, and its structural basis has been a puzzling question.Currently there are three independent MTBD atomic structures in the Protein Data Bank (PDB): One of the crystal structures of the D. discoideum dynein motor domain contains the MTBD (Fig. 3 A), and Carter et al. (2008) crystallized the MTBD of mouse cytoplasmic dynein fused with a seryl tRNA-synthetase domain (Fig. 3 C). The MTBD structure of C. reinhardtii axonemal dynein was solved using nuclear magnetic resonance (PDB accession no. 2RR7; Fig. 3 B). The MTBD is mostly composed of α-helices and the three structures are quite similar to each other within the globular MTBD (Fig. 3). Note that dynein c has an additional insert at the MTBD–microtubule interface (Fig. 3 B, inset), whose function is not yet clear. The three structures start to deviate from the junction between the MTBD and the coiled-coil region of the stalk (Fig. 3, A–C, blue arrowheads). Particularly, one of the stalk α-helix (CC2) in D. discoideum dynein motor domain appears to melt at the junction with the MTBD (Fig. 3 A, red arrowhead). This structural deviation suggests that the stalk coiled coil at the junction is flexible, which is consistent with the observation by EM (Roberts et al., 2009).Open in a separate windowFigure 3.Atomic models of the MTBD of dynein. (A) D. discoideum cytoplasmic dynein (PDB accession no. 3VKH). (B) C. reinhardtii dynein c (PDB accession no. 2RR7). The inset shows the side view, highlighting the dynein c–specific insert. (C) Mouse cytoplasmic dynein (PDB accession no. 3ERR). (D) Mouse cytoplasmic dynein fit to the MTBD–microtubule complex derived from cryo-EM (PDB accession no. 3J1T). All the MTBD structures were aligned using least square fits and color-coded with a gradient from the N to C terminus. CC1, coiled coil helix 1; CC2, coiled coil helix 2. The blue arrowheads points to the junction between MTBD and the stalk, where a well-conserved proline residue (colored pink) is located. In C and D, two residues (isoleucine 3269 and leucine 3417) are shown as spheres. The two residues form hydrophobic contacts in the β-registry (C), whereas they are separated in the α-registry (D) because of the sliding between the two α-helices (blue and red arrows). Conformational changes observed in the mouse dynein MTBD in complex with a microtubule by cryo-EM are shown by black arrows. Note that the cryo-EM density map does not have enough resolution to observe sliding between CC1 and CC2. The sliding was modeled based on targeted molecular dynamics (Redwine et al., 2012).Various mechanisms have been proposed to explain how the affinity between the MTBD and a microtubule is controlled. Gibbons et al. (2005) proposed “the helix-sliding hypothesis” (for review see Cho and Vale, 2012). In brief, this hypothesis proposes that the sliding between two α-helices CC1 and CC2 (Fig. 3, C and D; blue and red arrows) may control the affinity of this domain to a microtubule. When Gibbons’s classification (Gibbons et al., 2005) of the sliding state is applied to the three MTBD structures, the stalk in the D. discoideum dynein motor domain is in the “α-registry” state (not visible in Fig. 3 A because of the melting of CC2), which corresponds to the strong binding state. However, the mouse cytoplasmic and C. reinhardtii axonemal MTBDs have the “β-registry” stalk (Fig. 3 C), which corresponds to the weak binding state.To observe conformational changes induced by the α-registry and/or microtubule binding, Redwine et al. (2012) solved the structure of mouse dynein MTBD in complex with a microtubule at 9.7-Å resolution using cryo-EM and single particle analysis. The MTBD was coupled with seryl tRNA-synthetase to fix the stalk helix in the α-registry. At this resolution, α-helices are visible, and they used molecular dynamics to fit the crystal structure of mouse MTBD (β-registry) to the cryo-EM density map. According to this result, the first helix H1 moves ∼10 Å to a position that avoids a clash with the microtubule (Fig. 3 D, black arrows). This also induces opening of the stalk helix (CC1). Together with mutagenesis and single-molecule motility assays, Redwine et al. (2012) proposed that this new structure represents the strong binding state. Currently, it is not clear why the MTBD structure of D. discoideum dynein motor domain (α-registry, Fig. 3 A) is not similar to the new α-registry mouse dynein MTBD, and this problem needs to be addressed by further studies.

Structures around the first ATP binding site

Another central question about motor proteins is how Ångstrom-scale changes around the nucleotide are amplified to generate steps >8 nm. For dynein, the interface between the first nucleotide-binding pocket and the linker seem to be the key force-generating element (Fig. 4). The crystal structures of dynein give us clues about how nucleotide-induced conformational changes may be transmitted to and amplified by the linker domain.Open in a separate windowFigure 4.Structures around the first ATP binding site. (A) Schematic domain structure of the head domain. Regions contacting the linker domain are colored purple. (B) AAA submodules surrounding the first nucleotide-binding pocket (PDB accession no. 3VKG, chain A). The linker is connected to AAA1 domain by the “N-loop.” To highlight that the two finger-like structures are protruding, the shadow of the atomic structure has been cast on the plane parallel to the head domain. (C) Interaction between the linker and the two finger-like structures. The pink arrowhead points to the hinge-like structure of the linker. The pink numbers indicates the subdomain of the linker.The main ATP catalytic site is located between AAA1 and AAA2 (Fig. 4, A and B). There are four ADP molecules in the D. discoideum dynein crystal structures, but the first ATP binding site alone drives the microtubule-activated ATPase activity, based on biochemical experiments on dyneins whose ATP binding sites were mutated (Kon et al., 2012).One AAA+ module is composed of a large submodule and a small α submodule (Fig. 4 B). The large α/β submodule is located inside of the ring and the small α submodule is located outside. The large submodule bulges toward the linker face, and the overall ring forms a dome-like shape (Fig. 1 D).The main ATP catalytic site is surrounded by three submodules: AAA1 large α/β, AAA1 small α, and AAA2 large α/β (Fig. 4, A and B). Based on the structural changes of other AAA+ proteins (Gai et al., 2004; Suno et al., 2006; Wendler et al., 2012), the gap between AAA1 and AAA2 modules is expected to open and close during the ATPase cycle.In fact, the size of the gap varies among the dynein crystal structures. The crystal structures of yeast dyneins show a larger gap between AAA1 and AAA2, which might be the reason why no nucleotide was found in the binding pocket. Although Schmidt et al. (2012) soaked the crystals in a high concentration of various nucleotides (up to 25 mM of ATP), no electron densities corresponding to the nucleotide were observed at the first ATP binding site. Among dynein crystal structures, one of D. discoideum dynein (PDB accession no. 3VKH, chain A) has the smallest gap, but it is still considered to be in an “open state” because the arginine finger in the AAA2 module (Fig. 4 B, red) is far from the phosphates of ADP. Because the arginine finger is essential for ATP hydrolysis in other AAA+ proteins (Ogura et al., 2004), the gap is expected to close and the arginine finger would stabilize the negative charge during the transition state of ATP hydrolysis.The presumed open/close conformational change between AAA1 and AAA2 would result in the movement of two “finger-like” structures protruding from the AAA2 large α/β submodule (Fig. 4 B). The two finger-like structures are composed of the H2 insert β-hairpin and preSensor I (PS-I) insert. In D. discoideum dynein crystal structure, the two finger-like structures are in contact with the “hinge-like cleft” of the linker (Fig. 4 C, pink arrowhead). The hinge-like cleft is one of the thinnest parts of the linker, where only one α-helix is connecting between the linker subdomains 2 and 3.In the yeast crystal structures, which have wider gaps between AAA1 and AAA2, the two finger-like structures are not in direct contact with the linker and separated by 18 Å. Instead, the N-terminal region of the linker is in contact with the AAA5 domain (Fig. 4 A). To test the functional role of the linker–AAA5 interaction, Schmidt et al. (2012) mutated a residue involved in the interaction (Phe3446) and found that the mutation resulted in severe motility defects, showing strong microtubule binding and impaired ATPase activities. In D. discoideum dynein crystals, there is no direct interaction between AAA5 and the linker, which suggests that the gap between AAA1 and AAA2 may influence the interaction between the head and linker domain. The contact between the linker and AAA5 may also influence the gap around AAA5, because the gap between AAA5 and AAA6 is large in yeast dynein crystal, whereas the one between AAA4 and AAA5 is large in D. discoideum dynein.The movement of two finger-like structures would induce remodeling of the linker. According to the recent cryo-EM 3D reconstructions of cytoplasmic dynein and axonemal dynein c (Roberts et al., 2012), the linker is visible across the head and there is a large gap between AAA1 and AAA2 in the no-nucleotide state. This linker structure is considered to be the “straight” state (Fig. 2, A and E). In the presence of ADP vanadate, the gap between AAA1 and AAA2 is closed and the N-terminal region of linker is near AAA3, which corresponds to the pre-power stroke “hinged” state (Fig. 2, C and D). The transition from the hinged state to the straight state of the linker is considered to be the force-generating step of dynein.

Processivity of dynein

As the structure of dynein is different from other motor proteins, dynein’s stepping mechanism is also distinct. Both dynein and kinesin are microtubule-based motors and move processively. Based on the single molecule tracking experiment with nanometer accuracy (Yildiz et al., 2004), it is widely accepted that kinesin moves processively by using its two motor domain alternately, called the “hand-over-hand” mechanism. To test whether dynein uses a similar mechanism to kinesin or not, recently Qiu et al. (2012) and DeWitt et al. (2012) applied similar single-molecule approaches to dynein.To observe the stepping, the two head domains of yeast recombinant cytoplasmic dynein were labeled with different colors and the movement of two head domains was tracked simultaneously. If dynein walks by the hand-over-hand mechanism, the step size would be 16 nm and the stepping of one head domain would always be followed by the stepping of another head domain (alternating pattern), and the trailing head would always take a step (Fig. 2 F). Contrary to this prediction, both groups found that the stepping of the head domains is not coordinated when the two head domains are close together. These observations indicated that the chances of a leading or trailing head domain stepping are not significantly different (Fig. 2 G; DeWitt et al., 2012; Qiu et al., 2012).This stepping pattern predicts that the distance between the head domains can be long. In fact, the distance between the two head domains is on average ∼18 ± 11 nm (Qiu et al., 2012) or 28.4 ± 10.7 nm (DeWitt et al., 2012), and as large as ∼50 nm (DeWitt et al., 2012). When the two head domains are separated, there is a tendency where stepping of the trailing head is preferred over that of the forward head.In addition, even though the recombinant cytoplasmic dynein is a homodimer, the two heavy chains do not function equally. While walking along the microtubule, the leading head tends to walk on the right side, whereas the trailing head walks on the left side (DeWitt et al., 2012; Qiu et al., 2012). This arrangement suggests that the stepping mechanism is different between the two heads. In fact, when one of the two dynein heavy chains is mutated to abolish the ATPase activity at AAA1, the heterodimeric dynein still moves processively (DeWitt et al., 2012), with the AAA1-mutated dynein heavy chain remaining mostly in the trailing position. This result clearly demonstrates that allosteric communication between the two AAA1 domains is not required for processivity of dynein. It is likely that the mutated head acts as a tether to the microtubule, as it is known that wild-type dynein can step processively along microtubules under external load even in the absence of ATP (Gennerich et al., 2007).These results collectively show that dynein moves by a different mechanism from kinesin. It is likely that the long stalk and tail allow dynein to move in a more flexible manner.

Dyneins in the axoneme

As mentioned in the introduction, >10 dyneins work in motile flagella and cilia. The core of flagella and cilia is the axoneme, which is typically made of nine outer doublet microtubules and two central pair microtubules (“9 + 2,” Fig. 5 A). The axonemes are found in various eukaryotic cells ranging from the single-cell algae C. reinhardtii to human. Recent extensive cryo-electron tomography (cryo-ET) in combination with genetics revealed the highly organized and complex structures of axonemes that are potentially important for regulating dynein activities (Fig. 5, C and D; Nicastro et al., 2006; Bui et al., 2008, 2009, 2012; Heuser et al., 2009, 2012; Movassagh et al., 2010; Lin et al., 2012; Carbajal-González et al., 2013; Yamamoto et al., 2013).Open in a separate windowFigure 5.Arrangement of axonemal dyneins. (A) The schematic structure of the motile 9 + 2 axoneme, viewed from the base of flagella. (B) Quasi-planar asymmetric movement of the 9 + 2 axoneme typically observed in trachea cilia or in C. reinhardtii flagella. (C and D) 3D structure of a 96-nm repeat of doublet microtubules in the distal/central region of C. reinhardtii flagella (EMDB-2132; Bui et al., 2012). N-DRC, the nexin-dynein regulatory complex; ICLC, intermediate chain/light chain complex. Inner arm dynein subspecies are labeled according to Bui et al. (2012) and Lin et al. (2012). To avoid the confusion with the linker domain of dynein, the structures connecting between outer and inner arm dyneins are labeled as “connecters,” which are normally called “linkers.” Putative ATP binding sites of outer arm dynein determined by biotin-ADP (Oda et al., 2013) are indicated by orange circles. The atomic structure of cytoplasmic dynein is placed into the β-heavy chain of outer arm dynein and its enlarged view is shown in the inset. (D) Two doublet microtubules, viewed from the base of flagella.The basic mechanochemical cycles of axonemal dyneins are believed to be shared with cytoplasmic dynein. Dynein c is an inner arm dynein of C. reinhardtii and used extensively to investigate the conformational changes of dynein, as shown in Fig. 2 (A–E), by combining EM and single-particle analysis (Burgess et al., 2003; Roberts et al., 2012). Structural changes of axonemal dyneins complexed with microtubules are also observed by quick-freeze and deep-etch EM (Goodenough and Heuser, 1982; Burgess, 1995), cryo-EM (Oda et al., 2007), negative-staining EM (Ueno et al., 2008), and cryo-ET (Movassagh et al., 2010). According to these studies, the AAA+ head domains are constrained near the A-tubule in the no-nucleotide state. In the presence of nucleotide, the head domains move closer to the B-tubule and/or the minus end of microtubule, and their appearance becomes heterogeneous, which is consistent with the observation of isolated dynein c that shows greater flexibility between tail and stalk in the ADP/vanadate state (Burgess et al., 2003).One of the controversies about the structural changes of axonemal dyneins is whether their stepping involves “rotation” or “shift” of the head (Fig. 2, B to D). The stalk angle relative to the microtubule seems to be a constant ∼60° irrespective of the nucleotide state (Ueno et al., 2008; Movassagh et al., 2010). This angle is similar to the angle obtained from cryo-EM study of the MTBD–microtubule complex (Redwine et al., 2012). Based on these observations, Ueno et al. (2008) and Movassagh et al. (2010) hypothesize that the “shift” of the head pulls the B-microtubule toward the distal end. However, Roberts et al. (2012) propose that the “rotation” of head and stalk is involved in the stepping based on the docking of dynein c head into an averaged flagella tomogram obtained by Movassagh et al. (2010). This issue needs to be resolved by more reliable and high-resolution data, but these two models may not be mutually exclusive. For example, averaged tomograms may be biased toward the microtubule-bound stalk because tomograms are aligned using microtubules.To interpret these structural changes of axonemal dyneins, docking atomic models of dynein is necessary. According to Roberts et al. (2012), the linker face of inner arm dynein c is oriented outside of axoneme (Fig. 5 D). For outer arm dyneins, we used cryo-EM in combination with biotin-ADP-streptavidin labeling and showed that the ATP binding site, most likely AAA1, is on the left side of the AAA+ head (Fig. 5 C; Oda et al. (2013)). Assuming that the stalks extend out of the plane toward the viewer, the linker face of outer arm dynein is oriented outside of axoneme (Fig. 5 C, inset; and Fig. 5 D). If it were the opposite, the AAA1 would be located on the right side of the AAA+ head. In summary, both inner and outer arm dynein seem to have the same arrangement, with their linker face oriented outside of the axoneme (Fig. 5 D).A unique characteristic of axonemal dyneins is that these dyneins are under precise temporal and spatial control. To generate a planer beating motion (Fig. 5 B), dyneins should be asymmetrically controlled, because the dyneins located on doublets 2–4 drive the effective stroke, whereas the ones on doublets 6–8 drive the recovery stroke (Fig. 5 A). Based on the cryo-ET observation of axonemes, Nicastro et al. (2006) proposed that “linkers” between dyneins provide hard-wiring to coordinate motor activities. Because the linkers in axonemes are distinct structures from the linker domain of dynein, for clarity, here we call them “connecters.” According to the recent cryo-ET of proximal region of C. reinhardtii flagella (Bui et al., 2012), there are in fact asymmetries among nine doublets that are localized to the connecters between outer and inner arm dynein, called the outer-inner dynein (OID) connecters (Fig. 5, A and C). Recently we identified that the intermediate chain 2 (IC2) of outer arm dynein is a part of the OID connecters, and a mutation of the N-terminal region of IC2 affects functions of both outer and inner arm dyneins (Oda et al., 2013), which supports the idea that the connecters between dyneins are involved in axonemal dynein regulation.

Closing remarks

Thanks to the crystal structures, we can now design and interpret experiments such as single molecule assays and EM based on the atomic models of dynein. Our understanding of the molecular mechanism and cellular functions of dyneins will be significantly advanced by these experiments in the near future.One important direction of dynein research is to understand the motor mechanisms closer to the in vivo state. For example, the step sizes of cytoplasmic dynein purified from porcine brain is ∼8 nm independent of load (Toba et al., 2006). This result suggests that intermediate and light chain bound to the dynein heavy chain may modulate the motor activity of dynein. To address such questions, Trokter et al. (2012) reconstituted human cytoplasmic dynein complex from recombinant proteins, although the reconstituted dynein did not show robust processive movement. Further studies are required to understand the movement of cytoplasmic dynein. Similarly, axonemal dyneins should also be studied using mutations in a specific gene that does not affect the overall flagella structure, rather than depending on null mutants that cause the loss of large protein complexes.Detailed full chemomechanical cycle of dynein and its regulation are of great importance. Currently, open/closed states of the gap between AAA1 and AAA2 are not clearly correlated with the chemomechanical cycle of dynein. Soaking dynein crystal with nucleotides showed that the presence of ATP alone is not sufficient to close the gap, at least in the crystal (Schmidt et al., 2012). This result suggests that other factors such as a conformational change of the linker are required. For other motors, ATP hydrolysis is an irreversible chemical step, which is often “gated” by strain. In the case of kinesin, ATP is hydrolyzed by a motor domain only when a forward strain is applied by the other motor domain through the neck linker (Cross, 2004; Kikkawa, 2008). A similar strain-based gating mechanism may play important roles in controlling the dynein ATPase. Upon MTBD binding to the forward binding site, a strain between MTBD and tail would be applied to the dynein molecule. The Y-shaped stalk and strut/buttress under the strain would force the head domain to close the gap between AAA4 and AAA5 (Fig. 2 H). Similarly, the linker under the strain would be hooked onto the two finger-like structures and close the gap between AAA1 and AAA2 (Fig. 2 H). The gap closure then triggers ATP hydrolysis by dynein. This strain-based gating of dynein is consistent with the observation that the rate of nonadvancing backward steps, which would depend on ATP hydrolysis, is increased by load applied to dynein (Gennerich et al., 2007). To explain cilia and flagella movement, the geometric clutch hypothesis has been proposed (Lindemann, 2007), which contends that the forces transverse (t-force) to the axonemal axis act on the dynein to regulate dynein activities. In the axoneme, dynein itself can be the sensor of the t-force by the strain-based gating mechanism. Further experiments are required to test this idea, but the strain-based gating could be a shared property of biological motors.  相似文献   

3.
4.
Mitochondrial genes including Mfn2 are at the center of many diseases, underscoring their potential as a therapeutical target. The Chen group now identified 15-oxospiramilactone as a chemical inhibitor of the mammalian deubiquitylase USP30, acting on Mfn1 and Mfn2.Mitofusins, Fzo1 in yeast and Mfn1 and Mfn2 in mammals, are ubiquitylated and this post-translational modification has both positive and negative consequences on mitochondrial fusion1. The process of ubiquitylation requires enzymes belonging to three classes of proteins called E1, E2 and E3, which catalyze a cascade of successive steps leading to the covalent attachment of the modifier to its target protein2. Deubiquitylating enzymes render this modification reversible, thus offering further possibilities for regulation2. Ubiquitylation of mitofusins leads to their proteolyic breakdown, inhibiting fusion of mitochondria that consequently undergo fragmentation (Figure 1, left panel)1,3. For example in response to mitochondrial depolarization or apoptotic stimuli, E3 ligases like Parkin and Huwe1 ubiquitylate and target Mfn1 and Mfn2 to the proteasome (Figure 1, left panel)3,4. However, ubiquitylation of mitofusins is a dual process and a non-proteolytic role of mitofusin ubiquitylation that promotes mitochondrial fusion is now emerging1. This opposing mechanism was first described in yeast, where the isopeptidases Ubp12 and Ubp2 that deubiquitylate Fzo1 have been identified5. Inhibition and activation of mitochondrial fusion by ubiquitylation enable different morphologies of mitochondria ranging from a multitude of small organelles to a hyperconnected network (Figure 1)5. In a recent paper published in Cell Research, Yue et al.6 reveal that a similar process is present in mammalian cells. The authors report that the isopeptidase USP30 acts on ubiquitylated forms of Mfn1 and Mfn2 that stimulate mitochondrial fusion (Figure 1, right panel). This discovery identifies for the first time in mammals a positive role of ubiquitylation in the regulation of Mfn1 and Mfn2 fusion activity6.Open in a separate windowFigure 1Dual roles of ubiquitylation and deubiquitylation of mitofusins Mfn1 and Mfn2, the key effectors for mitochondrial fusion, in regulating mitochondrial fusion. On one hand, ubiquitylation of Mfn1 and Mfn2 by E3 ligases like Parkin or Huwe1 targets their proteasomal degradation and inhibits mitochondrial fusion, which results in mitochondrial fragmentation due to unopposed fission events. On the other hand, ubiquitylation of Mfn1 and Mfn2 by an unknown E3 ligase enhances their activity and promotes mitochondrial fusion. This positive regulation is counteracted by the deubiquitylase USP30, targeted by the small molecule inhibitor 15-oxospiramilactone.Moreover, Yue et al.6 identified the first small molecule inhibitor of mitochondrial fusion, 15-oxospiramilactone, which targets USP30 in both human and mouse cell lines. 15-oxospiramilactone is a semi-synthetic diterpene alkaloid of 330 Da that can be chemically synthetized through an oxidation reaction from spiramines extracted from the roots of a Chinese herbal medicine Spiraea japonica (Rosaceae). Inhibition of USP30 increased ubiquitylation of Mfn1 and Mfn2 and led to an elongation of the mitochondrial network (Figure 1, right panel)6,7. USP30 is a cysteine ubiquitin isopeptidase N-terminally anchored to the outer membrane of mitochondria, which was previously shown to regulate mitochondrial morphology dependent on Mfn1 and Mfn27. USP30 knockdown leads to mitochondrial elongation, a phenotype rescued by ectopic expression of wild-type USP30, while the catalytically inactive mutant C77S USP30 failed to revert7. Yue et al.6 show that 15-oxospiramilactone directly interacts with USP30, which also depends on its catalytically active cysteine, and inhibits the DUB activity of USP30 on tetraubiquitin chains. Moreover, they demonstrate that inhibition of USP30 and subsequent mitochondrial elongation are due to stimulated mitochondrial fusion activity, apparently with no influence on mitochondrial fission6. Concomitantly, cells showed increased ubiquitylation of Mfn1 and Mfn2 without significant changes in protein turnover of these two proteins6. Therefore, in analogy to findings in yeast, ubiquitylation of Mfn1 and Mfn2 can either signal them to activate mitochondrial fusion or in contrast promote their proteasomal degradation, resulting in mitochondrial fission (Figure 1).Importantly, 15-oxospiramilactone reverts the mitochondrial fragmentation phenotype of single Mfn-knockout (Mfn1−/− or Mfn2−/−) cells, suggesting that mitochondrial fusion depends on the ubiquitylation of both mitofusin proteins6. In yeast, the importance of ubiquitylation was proven by directly attaching a deubiquitylase to Fzo1, which resulted in a non-ubiquitylated and non-functional Fzo1 protein5. In addition, the identification and the subsequent mutagenesis study of the ubiquitylation sites in Fzo1 confirmed an interplay between ubiquitylation and oligomerization in mitochondrial fusion in S. cerevisiae5. Impairing the yeast E3 ligase SCFMdm30 inhibited mitochondrial fusion and, conversely, ablation of UBP12 led to more fusion events5,8. Given this new identification of USP30 as the functional orthologue of the yeast Ubp12, future studies will certainly aim at the identification of the E3 ligase counterpart of SCFMdm30 and ubiquitylation sites in Mfn1 and Mfn2. In addition to USP30 inhibition, other conditions leading to mitochondrial hyperfusion have been previously observed, such as mild stress conditions that increase reactive oxygen species (ROS)9. Importantly, oxidative stress and mitochondrial fusion are directly linked as ROS induces disulphide switching of Mfn2 to oligomeric forms that promote mitochondrial fusion9. It would be interesting to investigate whether 15-oxospiramilactone also affects the generation of disulphide-mediated mitofusin oligomers, thus activating mitochondrial fusion.Mutations in Mfn2 are causative for the Charcot-Marie-Tooth type 2A neuropathy, an autosomal dominant disorder of the peripheral nervous system that mainly affects axons and lower extremities1. Deficiencies in Parkin and Mfn2 ubiquitylation were also linked to Parkinson''s disease3. In addition to neuropathies, Mfn2 is associated to other diseases like cardiomyophathies and diabetes1. Yue et al.6 found that 15-oxospiramilactone reverted phenotypes arising from the lack of Mfn1 or Mfn2. It restored the normal distribution of mtDNA, allowed recovery of the ΔΨm and increased the ATP levels and OXPHOS capacity of the rebuilt mitochondrial network. Therefore, this study potentiates 15-oxospiramilactone for therapeutical benefit. The anti-cancer properties of 15-oxospiramilactone, also named S3 or NC043, have been previously reported10,11. It inhibits Wnt/β-catenin signaling and colon cancer cell tumorigenesis in a xenograft model10. Moreover, 15-oxospiramilactone increases Bim expression and apoptosis to inhibit tumor growth from Bax−/−/Bak−/− cells implanted in mice11. However, Yue et al.6 show that the effect of 15-oxospiramilactone in mitochondrial fusion is independent of apoptosis and suggest that the difference is due to drug concentration. Indeed, previous anti-cancer studies used 15-oxospiramilactone at a concentration range of 3.75-15 μM10,11, whereas 2 μM suffice to inhibit USP306. Further studies are needed to address the clinical relevance of 15-oxospiramilactone and USP30 in Mfn2-associated diseases.  相似文献   

5.
Relaxation of a hERG K+ channel model during molecular-dynamics simulation in a hydrated POPC bilayer was accompanied by transitions of an arginine gating charge across a charge transfer center in two voltage sensor domains. Inspection of the passage of arginine side chains across the charge transfer center suggests that the unique hydration properties of the arginine guanidine cation facilitates charge transfer during voltage sensor responses to changes in membrane potential, and underlies the preference of Arg over Lys as a mobile charge carrier in voltage-sensitive ion channels.The response of voltage-sensitive ion channels to changes in membrane potential is mediated by voltage sensor domains (VSD) containing a transmembrane helical segment (S4) with a repeating motif of positively charged and hydrophobic amino acids (Fig. 1) (1,2). Changes in membrane potential drive the S4 helix through the membrane plane with the charged side chains (largely arginine) on S4 swapping Glu/Asp carboxylate partners that lie on less mobile elements of the VSD (2). Movement of S4 is coupled to the ion-conducting pore to transmit changes in membrane potential to channel gating (3).Open in a separate windowFigure 1Structures of the VSD of membrane domains before MD in a POPC bilayer. The S2 (pink) and S4 (blue) helices of the VSD of the hERG model (A) and Kv1.2/2.1 chimera structure (B) are highlighted. (C) Sequence alignment of S2 and S4 among homologous voltage-sensitive K+ channels.The VSD charge-pairing motif of K+ and Na+ channels is best represented in VSD states at zero membrane potential (S4 helix up) for which crystal structures exist for Kv1.2 (4), Kv1.2/2.1 chimera (5), and Nav channels (6,7). In these states, positively charged residues on the intra- and extracellular sections of the S4 helix are separated by a hydrophobic charge-transfer center (CTC) (1) or plug (8) containing a highly conserved Phe residue (Fig. 1). This plug restricts water incursion across the VSD, focusing the electric field across a narrow region near the bilayer center. In voltage-driven transitions between S4 down- and up-states, positively charged S4 side chains move across the CTC.The ether-à-go-go (eag) and eag-related family of voltage-sensitive K+ channels likely share similar charge pairing interactions with VSDs in other channels (9,10). However, eag VSDs contain an extra negative charge on S2 (underlined in Fig. 1 C) so that in hERG, Asp residues (D460 and D466) lie approximately one helical turn above and below the conserved charge-transfer center Phe (F463) (Fig. 1). This eag-specific motif might be expected to facilitate transfer of Arg side chains through the CTC and to stabilize the voltage sensor (VS) in the up state. We recently described an open state (VS-up) hERG model built on the crystal structure template of the Kv1.2/2.1 chimera and molecular-dynamics (MD) simulation of this model in a hydrated POPC bilayer (11). We have inspected an extended version of this simulation and identified transitions of a gating charge into the CTC despite the absence of a membrane potential change. These transitions are absent in equivalent MD simulations of the chimera structure in a POPC bilayer.Fig. 1 shows a single VS from starting structures of the hERG model and the chimera structure in a hydrated POPC bilayer, after restrained MD to anneal the protein-lipid interface (see Methods in the Supporting Material). Because the hERG model is constructed on the chimera structure according to the alignment in Fig. 1 the pattern of pairing between S4 charges and acidic VS side chains is equivalent in the hERG model and chimera structure.The arrangement of charge-paired side chains remains constant during MD in all subunits of the chimera (e.g., Fig. 2 E and see Fig. S2 in the Supporting Material). However, in two subunits of the hERG model the R534 side chain moves toward the extracellular side of the bilayer, sliding into the CTC to form a charge interaction with the extra Asp residue (D460 in hERG) that lies just above F463 (Fig. 2, AC). This transition is facilitated by changes in side-chain rotamers of R534 and F463 as the planar Arg guanidine group rotates past the F463 ring, and the availability of D460 as a counterion for the R534 guanidine (Fig. 2). Movement of an Arg guanidine past the Phe side chain of the CTC is similar to that described in steered MD of an isolated VS domain (12).Open in a separate windowFigure 2Movement of the R534 side chain across the CTC in chain a of the hERG model simulation (A). Similar transitions are observed in chains a and b (panels B and C), but not chains c (D) or d (not shown), where the R534 side chain remains close to D466. In all subunits of the Kv1.2/2.1 chimera simulation, charge pairing of the starting structure (Fig. 1B) was maintained throughout (e.g., panel E and see Fig. S2 in the Supporting Material). (Black and blue lines) Distances from the Arg CZ or Lys ε atom to the two O atoms, respectively, of Asp or Glu.Mason et al. (13) have shown, using neutron scattering, that the low charge density guanidine cation (Gdm+) corresponding to the Arg side chain is poorly hydrated above and below the molecular plane. This property may underlie the universal preference for Arg (over Lys) in voltage sensor charge transfer. Although the poorly-hydrated surfaces of Gdm+ interact favorably with nonpolar (especially planar) surfaces (14,15), Gdm+ retains in-plane hydrogen bonding (13). In the transition of R534 across the CTC, in-plane solvation of the guanidine side chain is provided initially by D466, D501, and water molecules below the CTC, and during and after the transition by D501 and D460 side chains and waters above the CTC (Fig. 3, A and B). Complete transfer of the R534 side chain across the CTC was not observed, but would be expected to involve movement of the guanidine group away from H-bonding distance with D501.Open in a separate windowFigure 3In-plane solvation of R534 guanidine in the charge transfer center during the hERG model MD (A). (Dotted lines) H-bond distances of <2.5 Å. The right-hand group consists of top-down (B) and end-on (C) views of the distribution of oxygen atoms around the side chain of hERG R534 at 20-ns intervals during MD (subunit a). (D) End-on view of equivalent atom distributions around the K302 side chain during the Kv1.2/2.1 chimera MD (subunit c). (Red spheres, water O; pink, Asp OD1 and OD2; purple:, Glu OE1 and OE2.)The atom distribution around the R534 side chain during MD (Fig. 3, B and C) conforms to the experimental Gdm+ hydration structure (13), with H-bonding to waters and side-chain Asp O atoms exclusively in the guanidine plane. The passage of Gdm+ through the CTC is facilitated by the hydrophobic nature of Gdm+ above and below the molecular plane (13), which allows interaction with the nonpolar groups (especially F463) in the CTC (Fig. 3 A and see Fig. S3). This contrasts with the solvation properties of the Lys amino group (e.g., K302 of the Kv1.2/2.1 chimera (Fig. 1), which has a spherical distribution of H-bonding and charge-neutralizing oxygen atoms (Fig. 3 D and see Fig. S4).To further test these interpretations, we ran additional MD simulations of the isolated hERG VS domain model and an R534K mutant in a hydrated POPC bilayer. Again, the R534 side chain entered the CTC in the wild-type model simulation whereas the K534 side chain did not (see Fig. S5). Inspection of the atom distributions in Fig. 3 D (and see Fig. S4) indicates that the pocket below the conserved Phe of the CTC is particularly favorable for a Lys side chain, with waters and acidic side chains that satisfy the spherical solvation requirements of the terminal amino group, and nonpolar side chains that interact with the aliphatic part of the side chain.The occurrence of transitions of the R534 side chain through the CTC in the hERG model, in the absence of a change in membrane potential, indicates a relaxation from a less-stable starting structure. However, the path of the R534 side chain provides useful molecular-level insight into the nature of charge transfer in voltage sensors. How do these observations accord with broader evidence of charge transfer in voltage-sensitive channels in general, and hERG in particular? Studies with fluorinated analogs of aromatic side chains equivalent to F463 of hERG or F233 of the chimera indicate the absence of a significant role for cation-π interactions involving the CTC aromatic group in K+ and Nav channels, although a planar side chain is preferred in some cases (1,16). In hERG, F463 can be replaced by M, L, or V with small effects on channel gating (17), indicating that the hERG CTC requires only a bulky nonpolar side chain to seal the hydrophobic center of the VS and allow passage of the Arg side chain through the CTC. Both absence of requirement for cation-π interactions, and accommodation of nonplanar hydrophobic side chains in a functional hERG CTC, are broadly consistent with the interpretation that it is the poorly-hydrated nature of the Arg guanidine group above and below the molecular plane (together with its tenacious proton affinity (18)) that governs its role in carrying gating charge in voltage sensors.While the simulations suggest that R534 may interact with D460 in the open channel state, the possibility that the extra carboxylate side chain above the CTC might facilitate gating charge transfer is seemingly inconsistent with the slow activation of hERG, although hERG D460C does activate even more slowly than the WT channel (9). However, S4 movement in hERG occurs in advance of channel opening (19), and slow gating is partly mediated by interactions involving hERG cytoplasmic domains (20); thus, slow S4 movement may not be an inherent property of the hERG voltage sensor. Recent studies show that when hERG gating is studied at very low [Ca2+] (50 μM) and low [H+] (pH 8.0), the channel is strongly sensitized in the direction of the open state; this effect is reduced in hERG D460C (and hERG D509C) (10). These observations support a role for the extra hERG Asp residues in binding Ca2+ (and H+) (10), allowing the channel to be allosterically responsive to changes in pH and [Ca2+]. A true comparison of a hERG model with experimental channel gating might involve studies on a channel lacking cytoplasmic domains that modulate gating, and using conditions (high pH and low [Ca2+]) that leave the eag-specific Asp residues unoccupied. This could reveal the inherent current-voltage relationships and kinetics of the hERG voltage sensor.  相似文献   

6.
7.
8.
Division plane specification in animal cells has long been presumed to involve direct contact between microtubules of the anaphase mitotic spindle and the cell cortex. In this issue, von Dassow et al. (von Dassow et al. 2009. J. Cell. Biol. doi:10.1083/jcb.200907090) challenge this assumption by showing that spindle microtubules can effectively position the division plane at a distance from the cell cortex.Cell division, or cytokinesis, is accomplished via constriction of an equatorially localized contractile ring composed of filamentous actin and myosin II (Rappaport, 1996). Accurate division plane specification is essential to properly partition the cytoplasm and permit each daughter cell to receive a single copy of the genome. To ensure this accuracy, microtubules of the mitotic spindle signal to the cell cortex upon anaphase onset and promote assembly of the contractile ring between the separating chromosomes. The precise mechanism by which microtubules position the contractile ring, however, remains elusive.Early models on the nature of the spindle-derived signal proposed that astral rays (later found to be microtubules) position the division plane by either locally promoting contractility at the cell equator or inhibiting contractility at the cell poles (Rappaport, 1996). Recent evidence, though, suggests that distinct microtubule populations within a single cell provide multiple signals to promote accurate division (Canman et al., 2003; Bringmann and Hyman, 2005; Chen et al., 2008; Foe and von Dassow, 2008; von Dassow, 2009).The anaphase mitotic spindle contains several subtypes of microtubules, each of which is likely to contribute to division plane specification. Although kinetochore microtubules drive chromosome segregation during anaphase, nonkinetochore microtubules extend and maintain close proximity with the assembling central spindle (Mastronarde et al., 1993). Central spindle microtubules are highly stable (Salmon et al., 1976) and organize into an antiparallel bundled array between the separating chromosomes (Mastronarde et al., 1993). Preventing central spindle assembly usually results in a complete failure in cytokinesis, and prevents division plane specification in many cell types (Glotzer, 2005). Astral microtubules, however, are highly dynamic and grow out circumferentially from the centrosomes toward the cell cortex. Increasing evidence suggests that the astral microtubule signal inhibits contractility (see below; Canman et al., 2000; Kurz et al., 2002; Lewellyn et al., 2009).Regardless of the mechanism of division plane specification via microtubules, nearly all current models depend on direct contact between microtubules of the mitotic spindle and the cell cortex. Most of these models were based on observations that at the time of division plane specification, astral microtubules contact the cell cortex in nearly all systems studied. Nonkinetochore and/or central spindle microtubules have also been proposed to deliver critical contractile signals to the cell equator (Murata-Hori and Wang, 2002; Canman et al., 2003; Somers and Saint, 2003; Verbrugghe and White, 2004; Lewellyn et al., 2009; Vale et al., 2009). Yet in many cell types (especially early embryos), central spindle microtubules are at some distance from the cell cortex during division plane specification. Despite this, signal delivery for both astral and central spindle microtubules was proposed to occur via direct transport along microtubules to the cell cortex. The study of von Dassow et al. in this issue, however, indicates that accurate division plane specification does not require any close microtubule/cortical contact and may occur via a diffusion-based mechanism (see also Salmon and Wolniak, 1990).By treating echinoderm and Xenopus embryos with controlled levels of trichostatin A (TSA), which destabilizes acetylated dynamic microtubules via inhibition of the tubulin deacetylase HDAC6 (Matsuyama et al., 2002), von Dassow et al. (2009) were able to preferentially prevent astral microtubule growth while leaving central spindle microtubules intact. TSA treatment did not block anaphase onset or central spindle assembly, but resulted in the complete disruption of all direct microtubule contact with the cell cortex. Nevertheless, TSA-treated cells were able to undergo cytokinesis successfully (Fig. 1 A). The lack of astral microtubules in TSA-treated cells was also recapitulated by double centrosome ablation, and again the cells were able to undergo cytokinesis (Fig. 1 B). In both experiments, cytokinesis occurred in a timely manner, but the contractile ring was broader than in control cells. Together, these data suggest that spindle microtubules are sufficient to provide a diffusible stimulatory signal capable of defining the cell division plane without any direct contact with the cell cortex (von Dassow et al., 2009).Open in a separate windowFigure 1.Testing models of division plane specification by targeting distinct microtubule populations. By selectively eliminating astral microtubules with either controlled TSA-treatment (A) or by double centrosome ablation (B), von Dassow et al. (2009) provide strong evidence that microtubule contact with the cell cortex is not essential for successful cytokinesis. When a single centrosome was ablated, the division plane was displaced away from the ablated aster (B); this suggests that astral microtubules provide an inhibitory signal. Further, anucleate cells would only complete cytokinesis if the intracentrosomal distance exceeded the distance from the centrosomes to the cell cortex (C).The authors noticed that cytokinesis occurred selectively at a position with reduced microtubule density in control cells; therefore, they explored the role of astral microtubules in division plane positioning. By selectively ablating one centrosome just before anaphase onset, von Dassow et al. (2009) were also able to provide strong support for an inhibitory role of astral microtubules in division plane specification. When a single centrosome was ablated, the division plane was displaced away from the remaining astral microtubules and toward the ablated centrosome (Fig. 1 B). Further evidence for an inhibitory role of astral microtubules in cytokinesis came from close examination of the intracentrosomal distance in anucleate cells that were able to undergo cytokinesis relative to those that were not. Cells were only able to undergo cytokinesis when the intracentrosomal distance exceeded the distance from the centrosomes to the cell cortex (Fig. 1 C), which suggests that cytokinesis requires an aster-free zone. The authors propose a mechanism in these anucleate cells whereby global activation of contractility drives division plane specification refined by a zone of astral separation (von Dassow et al., 2009). However, one possibility is that a central spindle still forms in these anucleate cells and thus provides the same diffusion-based signal that promotes division in cells without asters. Indeed, antiparallel arrays of bundled microtubules that resemble the central spindle are known to form between asters without intervening chromosomes in other systems (Savoian et al., 1999).To summarize, the results described by von Dassow et al. (2009) support a model in which central spindle microtubules provide a diffusible stimulatory signal to promote the assembly of a broad contractile ring, which is then refined by astral microtubules into a tight contractile ring. It is tempting to speculate on the molecular nature of this diffusible signal and mechanism of the astral refinement during cytokinesis. Signaling via the small GTPase Rho is required for cytokinesis and is dependent on spindle microtubules (Bement et al., 2005; Piekny et al., 2005). von Dassow et al. (2009) showed that in TSA-treated cells lacking astral microtubules, the equatorial zone of active Rho GTPase is broader relative to control cells. Rho activation is promoted (at least in part) via the central spindle–localized GTP exchange factor, ECT2 (Glotzer, 2005). In parallel, the GTPase-activating protein (GAP) CYK4/MgcRacGAP also associates with the central spindle, where it acts to both limit the zone of Rho activity (Miller and Bement, 2009) and to promote the inactivation of another small GTPase, Rac (D''Avino et al., 2004; Yoshizaki et al., 2004; Canman et al., 2008). Perhaps in parallel to central spindle mediated activation of Rho signaling, local inactivation of the inhibitory Rac signal via CYK4 GAP activity would further specify the division plane, even at a distance (Fig. 2). When the dynamic asters are present, they could then additionally amplify Rac signaling at the cell poles via a similar mechanism to what occurs during cell motility (Wittmann and Waterman-Storer, 2001). This local feedback loop would reinforce the positive signal coming from the central spindle via Rho activation and could help delimit active Rho at the cell equator (Fig. 2). Certainly, understanding how Rho activation can be propagated to the cell cortex via diffusion in such an accurate manner will be a major future challenge.Open in a separate windowFigure 2.Model for central spindle–mediated signaling via Rho family small GTPases. Central spindle–localized guanine nucleotide exchange factor ECT2 leads to Rho activation at the cell equator. At the same time, central spindle–localized CYK4 (a Rho family GAP) would also locally inactivate the inhibitory Rac signal. Further refinement of the zone of active Rho by astral microtubule–activated Rac could then sharpen the Rho zone into a tight contractile ring.  相似文献   

9.
10.
FtsZ, a bacterial homolog of eukaryotic tubulin, assembles into the Z ring required for cytokinesis. In Escherichia coli, FtsZ interacts directly with FtsA and ZipA, which tether the Z ring to the membrane. We used three-dimensional structured illumination microscopy to compare the localization patterns of FtsZ, FtsA, and ZipA at high resolution in Escherichia coli cells. We found that FtsZ localizes in patches within a ring structure, similar to the pattern observed in other species, and discovered that FtsA and ZipA mostly colocalize in similar patches. Finally, we observed similar punctate and short polymeric structures of FtsZ distributed throughout the cell after Z rings were disassembled, either as a consequence of normal cytokinesis or upon induction of an endogenous cell division inhibitor.The assembly of the bacterial tubulin FtsZ has been well studied in vitro, but the fine structure of the cytokinetic Z ring it forms in vivo is not well defined. Super-resolution microscopy methods including photoactivated localization microscopy (PALM) and three-dimensional-structured illumination microscopy (3D-SIM) have recently provided a more detailed view of Z-ring structures. Two-dimensional PALM showed that Z rings in Escherichia coli are likely composed of loosely-bundled dynamic protofilaments (1,2). Three-dimensional PALM studies of Caulobacter crescentus initially showed that Z rings were comprised of loosely bundled protofilaments forming a continuous but dynamic ring (1–3). However, a more recent high-throughput study showed that the Z rings of this bacterium are patchy or discontinuous (4), similar to Z rings of Bacillus subtilis and Staphylococcus aureus using 3D-SIM (5). Strauss et al. (5) also demonstrated that the patches in B. subtilis Z rings are highly dynamic.Assembly of the Z ring is modulated by several proteins that interact directly with FtsZ and enhance assembly or disassembly (6). For example, FtsA and ZipA promote ring assembly in E. coli by tethering it to the cytoplasmic membrane (7,8). SulA is an inhibitor of FtsZ assembly, induced only after DNA damage, which sequesters monomers of FtsZ to prevent its assembly into a Z ring (9). Our initial goals were to visualize Z rings in E. coli using 3D-SIM, and then examine whether any FtsZ polymeric structures remain after SulA induction. We also asked whether FtsA and ZipA localized in patchy patterns similar to those of FtsZ.We used a DeltaVision OMX V4 Blaze microscope (Applied Precision, GE Healthcare, Issaquah, WA) to view the high-resolution localization patterns of FtsZ in E. coli cells producing FtsZ-GFP (Fig. 1). Three-dimensional views were reconstructed using softWoRx software (Applied Precision). To rule out GFP artifacts, we also visualized native FtsZ from a wild-type strain (WM1074) by immunofluorescence (IF).Open in a separate windowFigure 1Localization of FtsZ in E. coli. (A) Cell with a Z ring labeled with FtsZ-GFP. (B) Rotated view of Z ring in panel A. (C) Cell with a Z ring labeled with DyLight 550 (Thermo Fisher Scientific, Waltham, MA). (D) Rotated view of Z ring in panel C. (B1 and D1) Three-dimensional surface intensity plots of Z rings in panels B and D, respectively. (E) A dividing cell producing FtsZ-GFP. The cell outline is shown in the schematic. (Asterisk) Focus of FtsZ localization; (open dashed ovals) filamentous structures of FtsZ. Three-dimensional surface intensity plots were created using the software ImageJ (19). Scale bars, 1 μm.Both FtsZ-GFP (Fig. 1, A, B, and B1) and IF staining for FtsZ (Fig. 1, C, D, and D1) consistently localized to patches around the ring circumference, similar to the B. subtilis and C. crescentus FtsZ patterns (4,5). Analysis of fluorescence intensities (see Fig. S1, A and B, in the Supporting Material) revealed that the majority of Z rings contain one or more gaps in which intensity decreases to background levels (82% for FtsZ-GFP and 69% for IF). Most rings had 3–5 areas of lower intensity, but only a small percentage of these areas had fluorescence below background intensity (34% for FtsZ-GFP and 21% for IF), indicating that the majority of areas with lower intensity contain at least some FtsZ.To elucidate how FtsZ transitions from a disassembled ring to a new ring, we imaged a few dividing daughter cells before they were able to form new Z rings (Fig. 1 E). Previous conventional microscopy had revealed dynamic FtsZ helical structures (10), but the resolution had been insufficient to see further details. Here, FtsZ visualized in dividing cells by 3D-SIM localized throughout as a mixture of patches and randomly-oriented short filaments (asterisk and dashed oval in Fig. 1, respectively). These structures may represent oligomeric precursors of Z ring assembly.To visualize FtsZ after Z-ring disassembly another way, we overproduced SulA, a protein that blocks FtsZ assembly. We examined E. coli cells producing FtsZ-GFP after induction of sulA expression from a pBAD33-sulA plasmid (pWM1736) with 0.2% arabinose. After 30 min of sulA induction, Z rings remained intact in most cells (Fig. 2 A and data not shown). The proportion of cellular FtsZ-GFP in the ring before and after induction of sulA was consistent with previous data (data not shown) (1,11).Open in a separate windowFigure 2Localization of FtsZ after overproduction of SulA. (A) Cell producing FtsZ-GFP after 0.2% arabinose induction of SulA for 30 min. (B) After 45 min. (B1) Magnified cell shown in panel B. (C) Cell producing native FtsZ labeled with AlexaFluor 488 (Life Technologies, Carlsbad, CA) 30 min after induction; (D) 45 min after induction. (D1) Magnified cell shown in panel D. Scale bars, 1 μm. (Asterisk) Focus of FtsZ localization; (open dashed ovals) filamentous structures of FtsZ.Notably, after 45 min of sulA induction, Z rings were gone (Fig. 2, B and B1), replaced by numerous patches and randomly-oriented short filaments (asterisk and dashed ovals in Fig. 2), similar to those observed in a dividing cell. FtsZ normally rapidly recycles from free monomers to ring-bound polymers (11), but a critical concentration of SulA reduces the pool of available FtsZ monomers, resulting in breakdown of the Z ring (9). The observed FtsZ-GFP patches and filaments are likely FtsZ polymers that disassemble before they can organize into a ring.We confirmed this result by overproducing SulA in wild-type cells and detecting FtsZ localization by IF (Fig. 2, C, D, and D1). The overall fluorescence patterns in cells producing FtsZ-GFP versus cells producing only native FtsZ were similar (Fig. 2, B1 and D1), although we observed fewer filaments with IF, perhaps because FtsZ-GFP confers slight resistance to SulA, or because the increased amount of FtsZ in FtsZ-GFP producing cells might titrate the SulA more effectively.Additionally, we wanted to observe the localization patterns of the membrane tethers FtsA and ZipA. Inasmuch as both proteins bind to the same C-terminal conserved tail of FtsZ (12–14), they would be expected to colocalize with the circumferential FtsZ patches in the Z ring. We visualized FtsA using protein fusions to mCherry and GFP (data not shown) as well as IF using a wild-type strain (WM1074) (Fig. 3 A). We found that the patchy ring pattern of FtsA localization was similar to the FtsZ pattern. ZipA also displayed a similar patchy localization in WM1074 by IF (Fig. 3 B).Open in a separate windowFigure 3Localization of FtsA (A) and ZipA (B) by IF using AlexaFluor 488. (C) FtsA-GFP ring. (D) Same cell shown in panel C with ZipA labeled with DyLight 550. (C1 and D1) Three-dimensional surface intensity plots of FtsA ring from panel C or ZipA ring from panel D, respectively. (E) Merged image of FtsA (green) and ZipA (red) from the ring shown in panels C and D. (F) Intensity plot of FtsA (green) and ZipA (red) of ring shown in panel E. The plot represents intensity across a line drawn counterclockwise from the top of the ring around the circumference, then into its lumen. Red/green intensity plot and three-dimensional surface intensity plots were created using the software ImageJ (19). Scale bar, 1 μm.To determine whether FtsA and ZipA colocalized to these patches, we used a strain producing FtsA-GFP (WM4679) for IF staining of ZipA using a red secondary antibody. FtsA-GFP (Fig. 3 C) and ZipA (Fig. 3 D) had similar patterns of fluorescence, although the three-dimensional intensity profiles (Fig. 3, C1 and D1) reveal slight differences in intensity that are also visible in a merged image (Fig. 3 E). Quantitation of fluorescence intensities around the circumference of the rings revealed that FtsA and ZipA colocalized almost completely in approximately half of the rings analyzed (Fig. 3 F, and see Fig. S2 A), whereas in the other rings there were significant differences in localization in one or more areas (see Fig. S2 B). FtsA and ZipA bind to the same C-terminal peptide of FtsZ and may compete for binding. Cooperative self-assembly of FtsA or ZipA might result in large-scale differential localization visible by 3D-SIM.In conclusion, our 3D-SIM analysis shows that the patchy localization of FtsZ is conserved in E. coli and suggests that it may be widespread among bacteria. After disassembly of the Z ring either in dividing cells or by excess levels of the cell division inhibitor SulA, FtsZ persisted as patches and short filamentous structures. This is consistent with a highly dynamic population of FtsZ monomers and oligomers outside the ring, originally observed as mobile helices in E. coli by conventional fluorescence microscopy (10) and by photoactivation single-molecule tracking (15). FtsA and ZipA, which bind to the same segment of FtsZ and tether it to the cytoplasmic membrane, usually display a similar localization pattern to FtsZ and each other, although in addition to the differences we detect by 3D-SIM, there are also likely differences that are beyond its ∼100-nm resolution limit in the X,Y plane.As proposed previously (16), gaps between FtsZ patches may be needed to accommodate a switch from a sparse Z ring to a more condensed ring, which would provide force to drive ring constriction (17). If this model is correct, the gaps should close upon ring constriction, although this may be beyond the resolution of 3D-SIM in constricted rings. Another role for patches could be to force molecular crowding of low-abundance septum synthesis proteins such as FtsI, which depend on FtsZ/FtsA/ZipA for their recruitment, into a few mobile supercomplexes.How are FtsZ polymers organized within the Z-ring patches? Recent polarized fluorescence data suggest that FtsZ polymers are oriented both axially and circumferentially within the Z ring in E. coli (18). The seemingly random orientation of the non-ring FtsZ polymeric structures we observe here supports the idea that there is no strong constraint requiring FtsZ oligomers to follow a circumferential path around the cell cylinder. The patches of FtsZ in the unperturbed E. coli Z ring likely represent randomly oriented clusters of FtsZ filaments that are associated with ZipA, FtsA, and essential septum synthesis proteins. New super-resolution microscopy methods should continue to shed light on the in vivo organization of these protein assemblies.  相似文献   

11.
During mating, yeast cells must perforate their rigid cell walls at the right place to allow cell–cell fusion. In this issue, Dudin et al. (2015; J. Cell Biol. http://dx.doi.org/jcb.201411124) image mating fission yeast cells with unprecedented spatiotemporal resolution. The authors find that when mating cells come into contact, they form aster-like actin structures that direct cell wall remodeling precisely to the point of contact.At its core, sex is about the fusion of two haploid cells to form a diploid. For nonmotile cells like yeasts, that requires growth of mating projections to bridge the distance between the mating partners (Fig. 1 A). Yeast cells are protected from osmotic lysis by rigid cell walls, and growth of the mating projection involves local secretion of hydrolases that make the cell wall more elastic at the growing tip (Klis et al., 2006). As the wall expands, new components are added by synthases to maintain a continuous, unbroken wall. The process is orchestrated by a “cell wall integrity” signaling pathway, which monitors cell wall stress and delicately balances hydrolysis and synthesis to guarantee that no holes develop (Levin, 2011). But when it comes to mating, a hole must be made in both partners’ walls at the point of contact to allow cell–cell fusion. Precise positioning is key, as an off-center hole would lead to lysis. How is such precision achieved?Open in a separate windowFigure 1.Cell fusion during yeast mating: focus and communication. (A) Mating fission yeast cells grow projections toward each other and fuse at the point of contact. (B, left) Secreted hydrolases weaken the rigid cell wall to enable expansion, and rapidly diffuse away. (B, right) At a point of cell–cell contact, diffusional escape paths are longer, so hydrolases build up. (C) Focused delivery of secretory vesicles (ves) in mating budding yeast after contact. The image is adapted from Gammie et al. (1998), © The American Society for Cell Biology. (D) Actin cables during growth of the projection (left) and in the fusion focus (right). (E) Distribution of hydrolases and synthases in fusing cells. (F) The fusion focus forms first in the h mating partner and then in the h+ mating partner. CW, cell wall; PM, plasma membrane; N, nucleus; V, vacuole.An appealingly simple hypothesis—based on the observation that many hydrolases are secreted enzymes that can only transiently degrade the wall before diffusing away (Fig. 1 B)—is that when the mating projections come into contact, hydrolases from one partner would diffuse into the local wall of the other. Because diffusional escape paths are longer when cells are juxtaposed, hydrolases would be concentrated and make a hole only at the point of contact (Huberman and Murray, 2014). However, this purely geometrical effect cannot be the whole story, as classic genetic studies identified mutants of Saccharomyces cerevisiae that grew mating projections and achieved cell wall contact but failed to degrade the cell wall between mating partners (Kurihara et al., 1994). One set of mutants revealed that fusion requires especially high levels of pheromone secretion, which suggests that mating partners signal to each other to coordinate local wall remodeling (Brizzio et al., 1996). Elegant cytological analyses of another set of mutants have also suggested that vesicles delivering hydrolases are targeted precisely to the site of cell–cell contact (Fig. 1 C; Gammie et al., 1998). These inferences are strongly supported and expanded by a study in this issue (Dudin et al.), which provides a beautifully detailed characterization of mating in the distantly related fission yeast Schizosaccharomyces pombe.Using time-lapse microscopy and super-resolution imaging to monitor components of the actin cytoskeleton, Dudin et al. (2015) found that actin cables directed myosin V traffic to a broad zone at the tip of the growing mating projection. However, after cell–cell contact, actin cables were tightly focused toward a central “fusion focus” (Fig. 1 D). After focus formation, hydrolases were concentrated in a narrow region, whereas synthases were still distributed broadly (Fig. 1 E). The authors suggest that tightly focused myosin V–mediated delivery of hydrolases overwhelms the local synthases to make a hole in the central cell wall. In the surrounding wall, synthases counteract hydrolases to maintain cell wall integrity.How does the fusion focus form? A mating-specific formin, Fus1, became tightly localized to a small spot, where it presumably promoted focused actin polymerization and barbed-end anchoring (Dudin et al., 2015). Focus formation could arise from highly focused upstream signaling by formin regulators like Cdc42. Another possibility is suggested by the observation that, as also seen in budding yeast (Sheltzer and Rose, 2009), myosin V was required for focus formation. Thus, one could envision a positive feedback focusing mechanism in which formin-nucleated actin cables enable myosin V–mediated delivery of formins or their activators. Cells in which fusion focus formation was blocked by mutation of Fus1 or myosin V were unable to degrade juxtaposed cell walls and kept growing longer projections, attesting to the importance of the focus in enabling cell wall degradation.Why does the fusion focus only form upon cell–cell contact? The walls of the mating projections display mating type–specific agglutinins, which help mating partners stick to each other and might conceivably signal that contact has been established. Alternatively, focus formation might be triggered upon perception of a high-threshold pheromone concentration (Brizzio et al., 1996): pheromone levels would rise as the projections approach each other, and might be further increased after contact due to the same geometrical considerations discussed earlier for hydrolases.Intriguingly, Dudin et al. (2015) found that one of the mating partners, the h cell, always developed an actin fusion focus before the other, the h+ cell (Fig. 1 F). The basis for this asynchrony is unknown, but if the focus is indeed triggered by a threshold pheromone level, it could be that one pheromone crosses the threshold before the other. The h cells produce M-factor, whereas h+ cells produce P-factor. If P-factor were to accumulate more rapidly at the contact site, it might reach critical levels and trigger h cells to make their focus first. The ensuing more focused secretion of M-factor by the h cell might then trigger and correctly position focus formation by the h+ cell. Whatever the mechanism, the finding that one partner always focuses first makes it attractive to speculate that this asynchrony enables communication between mating partners that allows them to coordinate focus formation directly across from each other.  相似文献   

12.
13.
The T-cell actin cytoskeleton mediates adaptive immune system responses to peptide antigens by physically directing the motion and clustering of T-cell receptors (TCRs) on the cell surface. When TCR movement is impeded by externally applied physical barriers, the actin network exhibits transient enrichment near the trapped receptors. The coordinated nature of the actin density fluctuations suggests that they are composed of filamentous actin, but it has not been possible to eliminate de novo polymerization at TCR-associated actin polymerizing factors as an alternative cause. Here, we use a dual-probe cytoskeleton labeling strategy to distinguish between stable and polymerizing pools of actin. Our results suggest that TCR-associated actin consists of a relatively high proportion of the stable cytoskeletal fraction and extends away from the cell membrane into the cell. This implies that actin enrichment at mechanically trapped TCRs results from three-dimensional bunching of the existing filamentous actin network.The T-cell actin cytoskeleton is critical for proper antigen recognition by the mammalian adaptive immune system. During T-cell receptor (TCR) triggering by antigen peptides presented on major histocompatibility proteins (pMHCs) on the surfaces of antigen-presenting cells (APCs), the T-cell actin cytoskeleton adopts a pattern of centrosymmetric retrograde flow (1–3). This simultaneously promotes further TCR triggering (4) and rearranges various T-cell membrane proteins and their APC counterparts into an organized cell-cell interface termed the immunological synapse (IS) (5–7). During this process, TCRs form microclusters that move to the center of the IS in an actin-dependent manner (8,9). When engineered physical barriers interrupt the centripetal motion of TCR clusters, actin flow slows near the pinned microclusters, and the cytoskeletal network transiently accumulates and dissipates at the sites (10,11). The amplitude and duration of the induced cytoskeletal fluctuations are much greater than would be expected for a random distribution of independent objects, indicating that the actin in the local environment is coordinated. Whether this coordination arises from a rearrangement in the existing F-actin network or represents de novo polymerization of the cytoskeleton, as predicted by the association of TCRs with actin polymerizing factors (12), remains unclear. Here, we use a dual-probe cytoskeleton labeling approach that has previously been applied to distinguish between stable and dynamic populations of actin by exploiting the different relative affinities of monomeric actin and actin-binding proteins toward each population (13). This strategy reveals that TCR-associated actin is composed primarily of the stable cytoskeletal fraction and that local enrichment results from three-dimensional bunching of the existing filamentous actin network.Primary T cells from mice transgenic for the AND TCR were triggered using synthetic APCs consisting of supported lipid bilayers functionalized with pMHC and the integrin ligand intercellular adhesion molecule 1. Nanopatterned metal grids on the bilayer substrate acted as diffusion barriers that prevented lateral transport of TCR-pMHC complexes (14,15). Transient enrichment of actin at TCR clusters trapped at these barriers was visualized using fluorescent fusions of actin itself (mKate2-β-actin) and the F-actin binding domain of utrophin (EGFP-UtrCH). Such a dual-probe strategy theoretically allows for discrimination between different pools of actin: dynamic populations characterized by high polymerization and/or short filament fragments tend to be relatively better labeled by direct actin fusions whereas stable populations composed of longer filaments can support higher labeling by fluorescent fusions of F-actin binding proteins. This visualization method has been validated in Xenopus oocytes, where it distinguishes actin populations during wound healing (13). It has not been explicitly applied to T cells; however, simultaneous labeling of the Jurkat cell cytoskeleton using EGFP-actin and Alexa 568-phalloidin reveals distinct populations of actin consistent with the results expected from Xenopus (13,16).Our results show that the T-cell periphery is relatively enriched in mKate2-β-actin (Fig. 1 C, box 1), while EGFP-UtrCH dominates toward the center of the IS (Fig. 1 C, box 2). We infer from this probe distribution that the cytoskeleton at the T-cell periphery is composed of short fragments and is a site of active polymerization, whereas at the center of the IS, actin filaments are longer and predominantly stable. This is consistent with previous models of the T-cell actin network (3,16). An effective way to highlight each of these cytoskeletal regions is to consider the relative ratios of the two probes at each location. In this case, a high UtrCH/actin ratio corresponds to stable actin, and a high actin/UtrCH ratio corresponds to dynamic actin (Fig. 1 D). When T cells are treated with cytochalasin D, an inhibitor of actin polymerization, the overall UtrCH/actin ratio of the cell decreases as would be expected from a general decrease in polymerized actin (see Movie S7 and Movie S8 in the Supporting Material). However, it should be noted that photobleaching can also shift the UtrCH/actin ratio over time. We limit quantitative analysis of the ratio to its spatial gradients at a single time point, but such analysis is possible in systems that permit rigorous calibration for probe expression and photobleaching.Open in a separate windowFigure 1Ratiometric imaging of the cytoskeleton in live T cells distinguishes between dynamic and stable actin populations. (A) mKate2-β-actin, (B) EGFP-UtrCH, and (C) merged images of a triggered T cell show different actin pools. The cutouts in panel C correspond to (1) a region high in dynamic actin featuring short, polymerizing filaments and/or actin monomers and (2) a region with a stable actin population featuring longer filaments to which UtrCH can bind. (D) The UtrCH/actin ratio image highlights pools of relatively high UtrCH (red) or actin (blue). (Scale bars: 5 μm.)Actin enrichment at trapped TCR clusters incorporates both mKate2-β-actin (Fig. 2, A and C) and EGFP-UtrCH (Fig. 2, B and C). The relative UtrCH/actin ratio at these sites (Fig. 2 D, box 2) is quite high relative to nearby background areas (Fig. 2 D, box 1), indicating that the actin is derived primarily from the stable actin population.Open in a separate windowFigure 2Receptor-induced cytoskeletal enrichment at sites of pinned TCRs corresponds to a primarily stable actin fraction. (A) mKate2-β-actin, (B) EGFP-UtrCH, and (C) merged images of a triggered T cell interacting with a nanopatterned supported lipid bilayer show actin enrichment corresponding to putative sites of pinned TCRs. (D) The UtrCH/actin ratio is high at sites displaying actin enrichment, indicating a primarily stable actin fraction in (1) these regions compared to (2) nearby background areas. (Scale bars: 5 μm.)The three-dimensional distribution of TCR-associated actin was analyzed in dual-labeled live T cells using a spinning disk confocal microscope. The recordings show actin extending away from the cell membrane in the vicinity of trapped TCRs, while the rest of the actin cytoskeleton remains relatively flat (Fig. 3 and see Fig. S1 in the Supporting Material). These protrusions of actin away from the membrane surface are predominantly composed of stable, filamentous actin, as indicated by their relatively high UtrCH/actin ratio (Fig. 3 B).Open in a separate windowFigure 3Three-dimensional ratiometric imaging shows that actin enrichment extends away from the cell membrane. Single planes from (A) merged mKate2-β-actin and EGFP-UtrCH and (B) UtrCH/actin ratio three-dimensional stacks show actin enrichment at the cell membrane. Cutouts represent Z projections passing through sites of (1) enrichment and (2) nearby background regions. The color distribution in panel B is analogous to that in Figs. 1D and and22D, and is omitted for clarity. (Scale bar: 5 μm in the x axis only. Scale box: 1 μm.)Our interpretation of these results is that the filamentous actin network is relatively dense at sites of pinned TCRs. This is the simplest explanation out of several possibilities, one of which is formin-mediated mKate2-β-actin-deficient actin nucleation (17). Filament bunching at pinned TCRs can arise from consistent biophysical properties without assuming heterogeneity between the biochemistry of these receptors and other actin-associated proteins such as those at the cell edge, where locally high probe ratios are absent.Although TCRs are intentionally trapped as part of this experimental strategy, it is likely APCs can naturally impede TCR ligand mobilities under certain circumstances, and this has been shown to impact T-cell signaling (18,19). Actin architecture near cell surface proteins has been extensively studied in focal adhesions of fibroblasts (20), but the lack of stress fibers in T cells makes it unlikely that the two structures are similar. Thus, receptor-induced cytoskeletal enrichment at TCR clusters adds to the catalog of actin behaviors in situ, which is conveniently probed by techniques such as ratiometric dual-probe imaging in live cells. These techniques can be coupled to various spatial analysis algorithms to further extend their utility.  相似文献   

14.
Calmodulin regulation (calmodulation) of the family of voltage-gated CaV1-2 channels comprises a prominent prototype for ion channel regulation, remarkable for its powerful Ca2+ sensing capabilities, deep in elegant mechanistic lessons, and rich in biological and therapeutic implications. This field thereby resides squarely at the epicenter of Ca2+ signaling biology, ion channel biophysics, and therapeutic advance. This review summarizes the historical development of ideas in this field, the scope and richly patterned organization of Ca2+ feedback behaviors encompassed by this system, and the long-standing challenges and recent developments in discerning a molecular basis for calmodulation. We conclude by highlighting the considerable synergy between mechanism, biological insight, and promising therapeutics.The ancient Ca2+ sensor protein calmodulin (CaM) has emerged as a pervasive modulator of ion channels (Saimi and Kung, 2002), manifesting remarkable Ca2+ sensing capabilities in this context (Dunlap, 2007; Tadross et al., 2008) and furnishing essential Ca2+ feedback in numerous biological settings (Alseikhan et al., 2002; Xu and Wu, 2005; Mahajan et al., 2008; Adams et al., 2010; Morotti et al., 2012). Such modulation was discovered via mutations in CaM that altered the motile behavior of Paramecium, stemming from blunted activation of Ca2+-dependent Na+ current or K+ current (Kink et al., 1990). Since then, numerous ion channels have been found to be modulated by CaM, as extensively reviewed (Budde et al., 2002; Saimi and Kung, 2002; Wei et al., 2003; Halling et al., 2006; Tan et al., 2011; Nyegaard et al., 2012). This paper focuses on the CaM regulation (calmodulation) of voltage-gated Ca2+ channels, which are extensively regulated by intracellular Ca2+ binding to CaM (Ca2+/CaM; Lee et al., 1999; Peterson et al., 1999; Zühlke et al., 1999; Haeseleer et al., 2000; DeMaria et al., 2001; Budde et al., 2002; Halling et al., 2006). Such feedback regulation by Ca2+/CaM demonstrates versatile functional capabilities, represents a prominent prototype for ion-channel modulation, and holds far-reaching biological consequences. These attributes contribute much to the standing of voltage-gated Ca2+ channels as the queen of ion channels, molecules that not only sculpt membrane electrical waveforms but also serve as gatekeepers of the ubiquitous second messenger Ca2+ (Yue, 2004).

Classic history of discovery

The earliest signs of Ca2+-dependent regulation of Ca2+ channels came from data showing that increased Ca2+ could accelerate the inactivation of Ca2+ currents in Paramecium (Brehm and Eckert, 1978), invertebrate neurons (Tillotson, 1979), and insect muscle (Ashcroft and Stanfield, 1981). These results gave birth to the concept of Ca2+-dependent inactivation (CDI), the then counterintuitive notion that entities other than transmembrane voltage could modulate the rapid gating of ion channels (Eckert and Tillotson, 1981).Methodological advances permitting routine isolation and recording of Ca2+ currents within vertebrate preparations clearly revealed the existence of CDI of cardiac L-type Ca2+ currents (carried by CaV1.2 channels) in both multicellular preparations (Kass and Sanguinetti, 1984; Mentrard et al., 1984) and isolated myocytes (Lee et al., 1985). Classic data illustrating CDI of cardiac L-type currents is shown in Fig. 1 (A–C). Fig. 1 A (Ca) shows Ca2+ current from frog cardiocytes, elicited by a test voltage pulse to +80 mV (Mentrard et al., 1984). CDI becomes apparent upon Ca2+ entry during a voltage prepulse to +80 mV (Fig. 1 B), which causes sharply attenuated Ca2+ current in a subsequent test pulse (diminished area of red shading). To exclude voltage depolarization itself as the cause of inactivation, a prepulse to +120 mV (Fig. 1 C) can be demonstrated to produce far less inactivation within a subsequent test-pulse current. Because this prepulse imposed strong depolarization, but diminished Ca2+ entry via decreased driving force, the reemergence of test Ca2+ current suggests that the strong inactivation evident in Fig. 1 B could be attributed to CDI. Data of this type specifies an inactivation mechanism that depends on voltage as a U-shaped function, now considered a hallmark of CDI (Eckert and Tillotson, 1981).Open in a separate windowFigure 1.Classic U-shaped signature of CDI. (A–C) Early voltage-clamp recordings from a multicellular preparation of frog atrial trabeculae cells demonstrates CDI of Ca2+ currents. All listed voltages are relative to resting potential ER (∼−80 mV) Adapted from Mentrard et al. (1984). (A) Ca2+ currents (shaded pink) evoked in response to an +80-mV depolarizing test pulse show robust inactivation. Fast capacitive transient was estimated by blocking these Ca2+ currents with 3 mM Co2+ solution (Co). Vertical bar corresponds to 0.5 µA of current; horizontal bar, 100 ms. (B) The Ca2+ current in response to the test pulse is sharply attenuated when preceded by an +80 mV prepulse. This reduction in current amplitude reflects the inactivation of Ca2+ channels. Format is as in A. (C) Further increase in the prepulse potential to +120 mV restores Ca2+ current amplitude (compare with B). Here, the diminished Ca2+ entry during the prepulse was insufficient to trigger CDI. The format is as in A. (D–F) Single channel recordings of L-type Ca2+ channels from adult rat ventricular myocytes also exhibit U-shaped dependence of Ca2+ channel inactivation. Adapted from Imredy and Yue (1994) with permission from Elsevier. (D) Depolarizing voltage pulse to +20 mV evokes representative elementary Ca2+ currents. Ensemble average currents are shown on the bottom. (E) When depolarizing pulse was preceded by +40 mV prepulse, the elementary Ca2+ currents are sparser and the first opening is delayed. This reduction in open probability parallels the sharp attenuation of macroscopic Ca2+ currents seen in B. The ensemble average is shown on the bottom. (F) Further increase in prepulse voltage to +120 mV led to a reversal of this gating pattern, once again highlighting the U-shaped dependence of CDI. Ensemble average is shown on the bottom.Still, the actual nature of effects of Ca2+ upon gating remained unclear. Fig. 1 (D–F) reproduces the profile of CDI at the single-molecule level (Yue et al., 1990; Imredy and Yue, 1992, 1994). Resolving data such as these is technically challenging, owing to the diminutive size of unitary Ca2+ currents (∼0.3 pA) and the sub-millisecond gating timescale involved. These data demonstrate the existence of U-shaped inactivation in the gating of a single cardiac L-type Ca2+ channel fluxing Ca2+, as present in an adult rat ventricular myocyte (Imredy and Yue, 1994). These results established that Ca2+ influx of a single channel suffices to trigger CDI, and demonstrated the clear correspondence of single-channel and multicellular behavior (Fig. 1). Thus, CDI emerged as a legitimate molecular-level modulatory process.

Advent of Ca2+ channel calmodulation

The Ca2+ sensor mediating Ca2+ regulation of Ca2+ channels has long been a matter of debate, with proposals of direct binding of Ca2+ to the channel complex (Standen and Stanfield, 1982; Plant et al., 1983; Eckert and Chad, 1984), and of Ca2+-dependent phosphorylation and/or dephosphorylation of channels (Chad and Eckert, 1986; Armstrong, D., C. Erxleben, D. Kalman, Y. Lai, A. Nairn, and P. Greengard. 1988. Abstracts of papers presented at the forty-second annual meeting of the Society of General Physiologists. Abstr. 20). The road toward identification of the actual Ca2+ sensor began with the cloning and expression of recombinant Ca2+ channels, thereby enabling structure–function studies (Snutch and Reiner, 1992). This phase of discovery was initiated by a bioinformatic observation (Babitch, 1990), pointing out that the carboxy terminus of voltage-gated Ca2+ channels contains a region with weak homology to an EF hand Ca2+-binding motif (Fig. 2 A, EF1). After the recognition of EF1, the existence of a second EF hand–like motif became apparent (Fig. 2 A, EF2). This led to chimeric channel analysis (de Leon et al., 1995), wherein segments of the carboxy termini of L-type (CaV1.2) and R-type (CaV2.3) Ca2+ channels were swapped. These two types of channels exhibit strong and weak CDI, respectively, under conditions of strong intracellular Ca2+ buffering (Liang et al., 2003), making them advantageous for chimeric analysis. Results from these experiments revealed that the proximal third of the carboxy terminus (Fig. 2 A, CI region) was important for CDI, and that EF1 of CaV1.2 channels was essential for the strong CDI in these channels (de Leon et al., 1995). One possible explanation was that EF1 might be the Ca2+ binding site for CDI postulated previously (Standen and Stanfield, 1982; Plant et al., 1983; Eckert and Chad, 1984). However, mutations within the Ca2+ binding loop of EF1 failed to eliminate CDI (Zhou et al., 1997; Peterson et al., 2000), focusing the search for the Ca2+ sensor elsewhere.Open in a separate windowFigure 2.Calmodulation: the ingredients and the flavors. (A) Channel diagram illustrates overall arrangement of structural landmarks critical for CDI. The Ca2+-inactivation (CI) region, spanning ∼160 residues of the channel carboxy terminus, is highly conserved across CaV1/2 channel families and is elemental for CDI. The proximal segment (PCI) of the CI region includes the dual vestigial EF hand (EF) segments (shaded purple and blue-green). The IQ domain, a canonical CaM binding motif critical for CDI, is located just downstream of the PCI segment. The NSCaTE element in the amino terminus of CaV1.2/1.3 channels is an N-lobe Ca2+/CaM effector site. The CBD element (gray) in the carboxy terminus of CaV2 channels is a CaM binding segment thought to be critical for CDI. (B) The table outlines functional bipartition of CaM in CaV channels and the corresponding spatial Ca2+ selectivities. In CaV1.2 and CaV1.3 channels, both the C-lobe and N-lobe of CaM enable fast CDI with local Ca2+ selectivity. Latent CDI of CaV1.4 channels is revealed upon deletion of an autoinhibitory domain in the distal carboxy terminus. Throughout the CaV2 channel family, the N-lobe of CaM evokes slow CDI with global Ca2+ spatial selectivity. In CaV2.1 channels, the C-lobe of CaM supports ultra-fast facilitation (CDF) with local Ca2+ selectivity. No C-lobe triggered modulation has been reported for CaV2.2 and CaV2.3 channels.Targets of CaM binding themselves often resemble CaM, a bilobed molecule with each lobe composed of two EF hands (Jarrett and Madhavan, 1991). The dual vestigial EF hands in the carboxy terminus of channels (Fig. 2 A) may be seen as resembling a lobe of CaM, which suggests that CaM itself might be the Ca2+ sensor for CDI. Initial studies exploring this notion, however, showed that pharmacological inhibition of CaM did not eliminate CDI of L-type Ca2+ channels (Imredy and Yue, 1994; Zühlke and Reuter, 1998). Nonetheless, deletions within the CaV1.2 channel CI region identified an IQ motif (Fig. 2 A) as a critical element in CDI (Zühlke and Reuter, 1998), and mutagenesis of this IQ domain weakens CDI (Qin et al., 1999). As IQ motifs often bind Ca2+-free CaM (apoCaM; Jurado et al., 1999), the possibility that CaM acts as a CDI sensor reemerged. Definitive evidence came with studies involving a Ca2+-insensitive mutant form of CaM (CaM1234), in which point mutations within all four EF hands eliminate Ca2+ binding (Xia et al., 1998). If “preassociation” of apoCaM with target molecules were a prerequisite for subsequent regulation by Ca2+ binding to this apoCaM, then CaM1234 could act as a dominant negative to silence regulation, as seen for small-conductance Ca2+-activated K channels (Xia et al., 1998). Indeed, when CaV1.2 channels were coexpressed with CaM1234 or its analogues, CDI was ablated or strongly suppressed (Peterson et al., 1999; Zühlke et al., 1999). Additionally, apoCaM was found to bind to the CaV1.2 CI region, with critical dependence upon the IQ segment (Erickson et al., 2001; Pitt et al., 2001; Erickson et al., 2003). In retrospect, apoCaM preassociation with CaV1.2 channels would sterically protect CaM from pharmacological effects (Dasgupta et al., 1989), rationalizing the prior insensitivity of CDI to such small-molecule perturbation. In all, these data firmly substantiated CaM as the sensor for CaV1.2 channel Ca2+ regulation.Initially, it was believed that only CaV1.2 L-type channels (Fig. 2 B) were subject to CaM-mediated Ca2+ regulation (Zamponi, 2003). However, this system of calmodulation was gradually recognized to pertain to most of the CaV1 and CaV2 (but not CaV3) branches of the CaV channel superfamily (Liang et al., 2003; Fig. 2 B). P-type (CaV2.1) channels were found to be Ca2+ regulated (Lee et al., 1999), and a Ca2+/CaM binding site downstream of the IQ element (CBD; Fig. 2 A) was argued to be important. In this initial study, no role for CaM preassociation was recognized, and no role for the IQ domain was found. Moreover, Ca2+ regulation of CaV2.1 channels manifests as a facilitation of current (Ca2+-dependent facilitation; CDF), followed by a slowly developing CDI. Thus, it appeared possible that the Ca2+ regulation of CaV2.1 channels might diverge mechanistically from that of CaV1.2 channels. However, apoCaM was later found to preassociate with CaV2.1 channels (Erickson et al., 2001), Ca2+-insensitive mutant CaM molecules were found to eliminate Ca2+ regulation in CaV2.1 channels (DeMaria et al., 2001; Lee et al., 2003), and the IQ domain was determined to be structurally essential for this regulation (DeMaria et al., 2001; Lee et al., 2003; Kim et al., 2008; Mori et al., 2008). That said, the role of the CBD segment remains contentious, with some studies still arguing for this segment’s importance in CDI (Lee et al., 2003). In contrast, deletion of the entire carboxy terminus after the IQ domain (including the CBD element) completely spared CDF and CDI of CaV2.1 channels (DeMaria et al., 2001; Chaudhuri et al., 2005). Overall, CaV2.1 and CaV1.2 channels appear to be Ca2+ regulated by a largely conserved scheme.Soon thereafter, calmodulation was established for the remaining members of the CaV2 class of channels (Fig. 2 B, CaV2.2 and CaV2.3), which indicates that this modulatory system pertains throughout the CaV1-2 channel superfamily (Liang et al., 2003). Strong CaM-mediated CDI was found in CaV1.3 channels (Xu and Lipscombe, 2001; Shen et al., 2006; Yang et al., 2006); a latent capacity for CaM-mediated CDI was observed in CaV1.4 channels (Singh et al., 2006; Wahl-Schott et al., 2006); and potential indications of CaM-mediated regulation of CaV1.1 channels have been described (Stroffekova, 2008, 2011).A few more nuanced points nonetheless merit attention. First, some types of calmodulation are insensitive to strong intracellular Ca2+ buffering (e.g., CaV1.2 CDI), whereas others are not (CaV2.3 CDI; Fig. 2 B). This contrasting sensitivity enables the use of chimeric channels under increased Ca2+ buffering to identify key structural determinants (de Leon et al., 1995), despite the existence of calmodulation across the channel superfamily. Second, the mechanisms underlying CDF in CaV1.2 channels remain mysterious, as the CDF of native channels of the heart is weak to start, and attenuated by blockade of Ca2+ release from neighboring ryanodine receptor channels (RYRs; Wu et al., 2001). Additionally, CDF of CaV1.2 channels is strongly manifest only in recombinant channels containing point mutations within the IQ domain, and only when they are expressed in frog oocytes (Zühlke et al., 2000). Curiously, CDF is not observed in recombinant CaV1.2 channels expressed in mammalian cell lines (Peterson et al., 1999). In contrast, CDI of CaV1.2 channels is strong and universally observed across experimental platforms. Third, Ca2+/CaM-dependent dephosphorylation by calcineurin was initially suggested as a mechanism for CDI of Ca2+ channels (Chad and Eckert, 1986); however, this proposition has remained controversial as previously reviewed (Budde et al., 2002). Recently, calcineurin was found to preassemble with the CaV1.2 channel complex through the scaffolding protein AKAP79/150 bound to the distal carboxy terminus of the channel (Oliveria et al., 2007). Furthermore, two additional maneuvers were reported to impede CDI of these channels (Oliveria et al., 2012): the overexpression of a mutant AKAP79/150 incapable of binding calcineurin, and inhibition of calcineurin activity. However, others have found that direct inhibition of calcineurin has no effect on the CDI of L-type channels within multiple neuronal preparations (Branchaw et al., 1997; Victor et al., 1997; Zeilhofer et al., 1999). Moreover, deletion of the entire distal carboxy terminus (including the AKAP79/150 binding segment) of CaV1.2 channels preserves strong CDI (de Leon et al., 1995; Erickson et al., 2001; Crump et al., 2013). Similarly, short variants of CaV1.3 channels lacking the AKAP harboring distal carboxy termini (Marshall et al., 2011) also fully support CDI (Xu and Lipscombe, 2001; Shen et al., 2006; Yang et al., 2006; Bock et al., 2011). Altogether, it may be that calcineurin and AKAP79/150 are indirect and context-dependent modulators of CDI, rather than direct effector molecules (Budde et al., 2002).

Functional bipartition of CaM and selectivity for local/global Ca2+ sources

The calmodulatory mechanism for Ca2+ channels supports remarkable forms of Ca2+ decoding. This feature echoes earlier discoveries of “functional bipartition” of CaM in Paramecium (Kink et al., 1990; Saimi and Kung, 2002). Here, “under-excitable” behavioral strains had mutations only in the N-lobe of CaM, whereas “over-excitable” strains had mutations only in the C-lobe. Thus, one lobe of CaM can mediate signaling to one set of functions, whereas the other lobe signals to an alternative set of operations. It was unclear, however, whether this bipartition extended to mammals. The requirement that Ca2+ regulation of mammalian Ca2+ channels requires apoCaM preassociation permitted direct exploration of this question. Coexpressing channels with “half mutant” CaM molecules (CaM12 and CaM34, where only C- or N-terminal lobes, respectively, bind Ca2+) revealed that the individual lobes of CaM can evoke distinct components of channel regulation (Fig. 2 B). In most CaV1 channels, the C-lobe induces a kinetically rapid phase of CDI, whereas the N-lobe yields a slower component (Peterson et al., 1999; Yang et al., 2006; Dick et al., 2008). For CaV1.2 channels, early studies that utilized high intracellular Ca2+ buffering found only minimal N-lobe–mediated CDI (Peterson et al., 1999). However, when interrogated under low intracellular Ca2+ buffering, slow but recognizable N-lobe–mediated CDI of CaV1.2 channels emerges (Dick et al., 2008; Simms et al., 2013). Most strikingly, in CaV2.1 channels, the lobes of CaM produce opposing polarities of regulation: the C-lobe of CaM triggers a kinetically rapid CDF, whereas the N-lobe evokes a slower CDI (DeMaria et al., 2001; Lee et al., 2003). CaV2.2 and CaV2.3 channels manifest CDI triggered mainly by the N-lobe of CaM (Liang et al., 2003). Whether this bipartition orchestrates divergent classes of behavior in higher-order animals remains an intriguing and open question (Wei et al., 2003).Beyond simply splitting the Ca2+ signal, however, the calmodulation of mammalian Ca2+ channels revealed that the lobes of CaM can selectively decode Ca2+ in different ways. Hints as to this capability came from the differential sensitivity of calmodulation in various channels to Ca2+ buffering. Processes evoked by the C-lobe of CaM are invariably insensitive to introduction of strong Ca2+ buffering, whereas N-lobe–mediated processes in CaV2 channels can be eliminated by the same maneuver. Strong buffering would only spare Ca2+ signals near the cytoplasmic mouth of channels where strong point-source Ca2+ influx would overwhelm buffer capacity (Neher, 1986; Stern, 1992). This argues that local Ca2+ influx through individual channels triggers C-lobe signaling; in other words, the C-lobe of CaM exhibits a “local Ca2+ selectivity.” In contrast, a spatially global elevation of Ca2+ (present in the absence of strong Ca2+ buffering) is required for N-lobe signaling of CaV2 channels (DeMaria et al., 2001; Lee et al., 2003; Liang et al., 2003); thus, the N-lobe in this context exhibits a “global selectivity.” The existence of global selectivity is notable, given that local Ca2+ influx yields far larger Ca2+ increases near a channel than does globally sourced Ca2+ (Tay et al., 2012). One exception to this pattern is the local Ca2+ selectivity of the N-lobe component of CDI in CaV1.2/1.3 channels (see two paragraphs below). Corroboration of spatial Ca2+ selectivity is provided by the ability of single CaV1.2 channels to undergo CDI (Fig. 1, D–F). By definition, only a local Ca2+ source is present in the single-channel configuration; thus, the presence of CDI in this context indicates local Ca2+ selectivity. Additionally, single-channel records of CaV2.1 channels exhibit CDF (driven by C-lobe), but not CDI (triggered by N-lobe; Chaudhuri et al., 2007), which is consistent with the proposed differential selectivities for this channel. Fig. 2 B summarizes the arrangement of spatial Ca2+ selectivities according to the lobes of CaM.The mechanisms underlying these contrasting spatial Ca2+ selectivities can be interpreted as emergent behaviors of a system in which a lobe of apoCaM must transiently detach from a preassociation site before binding Ca2+, and where a Ca2+-bound lobe of CaM must associate with a channel effector site to mediate regulation (Tadross et al., 2008). When the slow Ca2+-unbinding kinetics of the C-lobe of CaM are imposed on this scenario, local Ca2+ sensitivity invariably results. In contrast, when the rapid Ca2+-unbinding kinetics of the N-lobe are interfaced with this architecture, global Ca2+ selectivity arises if the channel preferentially binds apoCaM versus Ca2+/CaM, as in the case of CaV2 channels (Fig. 2 B). When the channel preferentially interacts with Ca2+/CaM, local Ca2+ selectivity emerges, as in the context of CaV1 channels (Fig. 2 B). The relative roles of the affinities and kinetics of Ca2+ binding to the two lobes of CaM in mediating local and global Ca2+ selectivities have been considered at length elsewhere (Tadross et al., 2008).As an explicit example of how spatial Ca2+ selectivity of N-lobe regulation is specified, we consider the effects of an N-terminal spatial Ca2+-transforming element (NSCaTE) present only on the amino terminus of CaV1.2/1.3 channels (Fig. 2 A). NSCaTE is a Ca2+/CaM binding site whose presence enhances the aggregate channel affinity for Ca2+/CaM over apoCaM (Ivanina et al., 2000; Dick et al., 2008), endowing the N-lobe component of CDI of these channels with a largely local Ca2+ selectivity of N-lobe CDI in these channels. Elimination of the NSCaTE site in these channels, which tilts channels toward a preference for apoCaM, then switches their N-lobe CDI to a global profile. Conversely, donation of NSCaTE to CaV2 channels, which causes channels to favor Ca2+/CaM binding, then endows their N-lobe CDI with local Ca2+ selectivity (Dick et al., 2008). In this manner, the presence of NSCaTE can tune the spatial Ca2+ selectivity of N-lobe–mediated channel regulation. This overall arrangement of CaM interactions with a target molecule could endow numerous other regulatory systems with spatial Ca2+ selectivity.

Molecular basis of calmodulation

Elucidating the arrangement of apoCaM and Ca2+/CaM on Ca2+ channels is fundamental for the field, given the biological influence of calmodulation, and the importance of this system as an ion-channel regulatory prototype (Dunlap, 2007). This critical task, however, remains an ongoing challenge, given the >2,000 amino acids comprising the main pore-forming α1 subunit alone, the ability of multiple peptide segments of the channel to bind CaM in vitro, and the formidable nature of obtaining atomic structures for these channels.The stoichiometry of CaM interaction with channels has been debated. Crystal structures of Ca2+/CaM complexed with portions of the carboxy tail CI region of CaV1.2 channels (Fallon et al., 2009; Kim et al., 2010) suggest that multiple CaM molecules may interact to produce the full spectrum of Ca2+ regulatory functions. Moreover, CaM can interact with multiple peptide segments of the channel (Ivanina et al., 2000; Tang et al., 2003; Zhou et al., 2005; Dick et al., 2008; Ben Johny et al., 2013). In contrast, covalent fusion of single CaM molecules to CaV1.2 channels suggests that only one CaM per channel suffices to elicit CDI (Mori et al., 2004). Moreover, live-cell FRET studies of the interactions between CaM and holochannels (CaV1.2) also point to a 1:1 CaM/channel ratio (Ben Johny et al., 2012). In the holochannel, the various CaM binding segments may be arranged in close proximity so as to allow the two lobes of CaM to preferentially interact with the distinct channel segments during Ca2+ regulation (Dick et al., 2008; Ben Johny et al., 2013). Recent biochemical studies of the NSCaTE element show that this segment preferentially interacts with a single lobe of CaM (N-lobe) at a time (Liu and Vogel, 2012). Furthermore, the NSCaTE element can interact also with Ca2+/CaM prebound to an IQ domain peptide (Taiakina et al., 2013). Thus, we favor the interpretation that only one CaM interacts within holochannels, although peptide fragments of Ca2+ channels may bind to multiple CaM molecules.The IQ domain (Fig. 3 A, blue circle) is critical for calmodulation. Mutations within this domain markedly modulate the Ca2+ regulation of CaV1.2 channels (Zühlke and Reuter, 1998; Qin et al., 1999; Zühlke et al., 1999, 2000; Erickson et al., 2003), CaV1.3 channels (Yang et al., 2006; Bazzazi et al., 2013; Ben Johny et al., 2013), CaV2.1 channels (DeMaria et al., 2001; Lee et al., 2003; Kim et al., 2008; Mori et al., 2008), and CaV2.2 and CaV2.3 channels (Liang et al., 2003). Furthermore, apoCaM preassociation with CaV1.2/1.3 channels requires the IQ domain (Erickson et al., 2001; Pitt et al., 2001; Erickson et al., 2003; Bazzazi et al., 2013; Ben Johny et al., 2013). Finally, Ca2+/CaM binds well to IQ-domain peptides of many CaV channels (Peterson et al., 1999; Zühlke et al., 1999; DeMaria et al., 2001; Pitt et al., 2001; Liang et al., 2003; Bazzazi et al., 2013; Ben Johny et al., 2013). All these results gave rise to the IQ-centric hypothesis shown in Fig. 3 A. Here, the IQ domain is important for both apoCaM preassociation (left) and Ca2+/CaM binding (effector configuration shown on the right). This IQ-centric paradigm has motivated efforts to resolve crystal structures of Ca2+/CaM complexed with IQ-domain peptides of various CaV1-2 channels (Fallon et al., 2005; Van Petegem et al., 2005; Kim et al., 2008; Mori et al., 2008), as shown in Fig. 3 (B and C). Throughout, the IQ peptide segment appears as an α-helical entity with N and C termini as labeled.Open in a separate windowFigure 3.Toward an atomic-level understanding of CDI. (A) IQ-centric view of calmodulation of CaV channels. In this mechanistic scheme, ApoCaM is preassociated with the IQ domain (blue). Upon Ca2+ binding, CaM rebinds the same IQ domain with a higher affinity, and subtle conformational rearrangements are presumed to trigger CaM-mediated channel regulation. (B, left) Crystal structure of CaV1.2 IQ domain peptide in complex with Ca2+/CaM (Protein Data Bank accession no. 2BE6). The IQ domain is colored blue. CaM is show in cyan (N-lobe, pale cyan; C-lobe, cyan). Ca2+ ions are depicted as yellow spheres. The key isoleucine residue (red) serves as a hydrophobic anchor for CaM. Ca2+/CaM adopts a parallel arrangement with the IQ domain in which the N-lobe binds closer to the amino terminus of the IQ domain and the C-lobe binds further downstream. (B, right) Crystal structure of Ca2+/CaM bound to mutant CaV1.2 IQ domain with alanines substituted for the key isoleucine-glutamine residues (accession no. 2F3Z). This double mutation abolishes CDI. Structurally, however, Ca2+/CaM hugs the mutant IQ domain in a similar conformation as to its interaction with the wild-type (left). (C) Crystal structure of Ca2+/CaM bound to CaV2.1 IQ domain (left, accession no. 3BXK; right, accession no. 3DVM). The IQ domain is shaded green, CaM in cyan. Ca2+/CaM was reported to bind to the CaV2.1 IQ domain in both parallel (left) and antiparallel (right) arrangements. The antiparallel arrangement in which the C-lobe of CaM binds upstream of IQ domain has led to speculation that CDF may result from this inverted polarity of Ca2+/CaM association. (D) Simplified configurations of the CaM/channel complex relevant for calmodulation (E, A, IC, IN, and ICN). In configuration E, channels lack preassociated CaM and therefore cannot undergo CDI. Configuration A corresponds to channels bound to apoCaM, and thus capable of undergoing robust CDI. Ca2+ binding to the C-lobe and N-lobe of CaM leads to the inactivated configurations IC and IN, respectively. Ca2+ binding to both lobes of CaM then yields configuration ICN, with both C and N lobes of CaM engaged toward CDI. This final transition may exhibit positive cooperativity as specified by the constant λ >> 1. Under endogenous levels of CaM, channels may reside in any of the five configurations. Strong coexpression of wild-type or mutant CaM restricts accessibility to various states. Reproduced with permission from Ben Johny et al., 2013.However, this viewpoint remains problematic in three regards. (1) The atomic structures of Ca2+/CaM bound to wild-type and mutant IQ peptides of CaV1.2 (Fig. 3 B) show that a central isoleucine (side chains explicitly shown) in the IQ element is deeply buried within the C-lobe of Ca2+/CaM (Van Petegem et al., 2005), and that alanine substitution at this site hardly changes structure (Fallon et al., 2005). Additionally, Ca2+/CaM dissociation constants for corresponding wild-type and mutant IQ peptides are nearly the same (Zühlke et al., 2000; Bazzazi et al., 2013). Why then would alanine substitution at this well-ensconced site influence the rest of the channel to blunt regulation (Fallon et al., 2005)? (2) For CaV1.2/1.3 channels, the N-lobe of Ca2+/CaM effector site appears to be an NSCaTE element (Fig. 3 A, oval) of the channel amino terminus (Dick et al., 2008; Tadross et al., 2008; Liu and Vogel, 2012), separate from the IQ element. (3) Crystal structures of Ca2+/CaM in complex with the IQ peptide of CaV2.1 channels show that CaM can adopt both a parallel (Mori et al., 2008) and an antiparallel configuration (Kim et al., 2008; Fig. 3 C, left and right, respectively). The apparent inversion in configuration of CaM binding to IQ domain between CaV1.2 and CaV2.1 has been proposed as a mechanism for the opposing polarity of Ca2+ regulation observed in the two channels (Kim et al., 2008; Minor and Findeisen, 2010). However, detailed structural analysis along with functional systematic alanine scanning mutagenesis and chimeric channel analysis argues that the C-lobe effector site resides at a site beyond the IQ module (Mori et al., 2008).A major concern with older IQ domain analyses is that the regulatory system was not considered conceptually as a whole, as shown in Fig. 3 D (drawn with specific reference to CaV1.3 channels; Ben Johny et al., 2013). Configuration E portrays channels lacking apoCaM. Such channels can open normally, but do not manifest CDI because Ca2+/CaM from bulk solution cannot efficiently access a channel in configuration E to induce CDI (Mori et al., 2004; Yang, P.S., M.X. Mori, E.A. Antony, M.R. Tadross, and D.T. Yue. 2007. Biophysical Journal abstracts issue. 1669-Plat; Liu et al., 2010; Findeisen et al., 2011). ApoCaM binding with configuration E gives rise to configuration A, where opening can also occur normally, but CDI can now take place. For CDI, Ca2+ binding to both lobes of CaM yields configuration ICN, which underlies a fully inactivated channel with strongly reduced opening. For intermediate configurations, Ca2+ binding only to the C-lobe brings about configuration IC, equivalent to a C-lobe inactivated channel; Ca2+ binding only to the N-lobe yields the N-lobe–inactivated arrangement (IN). Of particular importance, ensuing entry into ICN likely involves positively cooperative interactions specified by cooperativity factor λ >> 1.This scheme emphasizes several challenges for older analyses of the IQ domain, where CDI was mostly assessed with only endogenous CaM present. Results thus obtained would be ambiguous, because IQ-domain mutations could alter calmodulation via changes at multiple steps depicted in Fig. 3 D, whereas other interpretations largely attributed the effects to altered Ca2+/CaM binding with an IQ effector site. In contrast, IQ mutations could well weaken apoCaM preassociation and reduce CDI by favoring configuration E. Moreover, functional deficits caused by mutations that do attenuate interaction with one lobe of Ca2+/CaM may be masked by positively cooperative steps (λ in Fig. 3 D).To minimize these issues, CDI of CaV1.3 channels was recently characterized during strong coexpression with various mutant CaM molecules (Bazzazi et al., 2013; Ben Johny et al., 2013) as shown in Fig. 4 (A–C, top). For orientation, CDI of channels expressed with only endogenous CaM present is shown in the upper portion of Fig. 4 A. Strong CDI produces a rapid decay of whole-cell Ca2+ current (red trace), compared with the negligible decline of Ba2+ current (black trace). Because Ba2+ binds CaM poorly (Chao et al., 1984), the decline of Ca2+ versus Ba2+ current after 300 ms of depolarization quantifies CDI (CDI parameter plotted below in bar graphs). One can isolate the diamond-shaped subsystem lacking configuration E (Fig. 4 B, top) by leveraging mass action with strong coexpression of wild-type CaM (CaMWT). Further simplification of CDI was obtained by strongly coexpressing channels with CaM12 (Fig. 4 C, top), which empties configuration E by mass action, and forbids configurations IN and ICN. Thus, the isolated C-lobe component of CDI can be studied, with the signature rapid time course of current decay shown near the top of Fig. 4 C. Critically, this layout avoids interplay with cooperative λ steps in Fig. 3 D. Likewise, strongly coexpressing CaM34 focuses on the slower N-lobe form of CDI, with attendant simplifications.Open in a separate windowFigure 4.Residue-level roadmap for calmodulation of CaV1.3 channels. Systematic alanine scanning mutagenesis of the entire CI domain of CaV1.3 channels. (A) CDI of wild-type and mutant CaV1.3 channels recombinantly expressed in HEK293 cells was measured with only endogenous CaM present. CDI observed here reflects properties of the entire system (stick figure) diagrammed in Fig. 3 D. Exemplar whole-cell current shows robust CDI for wild-type (WT) CaV1.3, reflecting their high affinity for apoCaM. Here and throughout, the vertical bar pertains to 0.2 nA of Ca2+ current (black); the Ba2+ current (gray) has been scaled ∼3-fold downward to aid comparison of decay kinetics. Horizontal bar, 100 ms. CDI is measured as the fractional reduction of Ca2+ current in comparison to Ba2+ at 300 ms (Ben Johny et al., 2013). Bottom, bar graph summary of CDI for alanine substitutions for indicated residues. CDI for WT channels, blue-green vertical line. Alanine substitutions in both EF hand regions and IQ domain resulted in diminished CDI. (B) Overexpression of CaMWT isolates the behavior of the diamond-shaped subsystem. Such overexpression of CaMWT should rescue CDI for mutations that weakened apoCaM preassociation. For WT CaV1.3, exemplar current shows that CDI is unaltered, as expected for a channel with high affinity for apoCaM. Bottom, bar graph summary of CDI for various mutant channels with CaMWT overexpressed. Format is as in A. CDI is rescued for several mutations in the EF hand region (EF2) and the IQ domain, reflecting the preassociation of N-lobe and C-lobe of apoCaM. (C) Overexpression of CaM12 isolates C-lobe CDI. Exemplar currents for WT channels show the fast C-lobe form of CDI. Bottom, bar graph summary shows the footprint of C-lobe CDI. Mutants in both the first EF hand and the IQ domain disrupted the C-lobe CDI (see the LGF loci in the EF hand regions and TFL and IQD loci in the IQ domain). The format is as in A. (D) Overexpression of CaM34 isolates N-lobe CDI. N-lobe CDI was largely preserved by mutations in the CI region. Mutations that weakened N-lobe apoCaM preassociation exhibited enhanced N-lobe CDI (see EF2 segment). Reassuringly, alanine scanning mutagenesis of NSCaTE element resulted in specific disruption of N-lobe CDI of CaV1.3 channels (Tadross et al., 2008). Adapted with permission from Ben Johny et al. (2013) and Bazzazi et al. (2013).Accordingly, a new framework of CaM/channel configurations that underlie calmodulation could be deduced, as diagrammed in Fig. 5 (Ben Johny et al., 2013). For convenience, Fig. 4 compiles results from a systematic alanine scan covering the entire CI domain of the carboxyl tail of CaV1.3 channels (Bazzazi et al., 2013; Ben Johny et al., 2013), where substitution positions are denoted to the left of the bar graph in Fig. 4 A. The N-terminal end of the CI segment corresponds to the top of the graph, purple and blue-green regions indicate EF1 and EF2, and the lavender zone denotes the IQ domain. Electrophysiological characterization for each of the substitutions was performed for all the various subsystems (Fig. 4, A–D, top), and bar graphs plot the strength of CDI for the corresponding subsystems in each of the panels. The blue-green vertical lines reference the profile for wild-type channels. The results thus obtained were combined with CaM binding assays (not depicted) to identify meaningful structure–function correlations.Open in a separate windowFigure 5.Next-generation blueprint for CaM/CaBP regulation of CaV channels. (A) De novo molecular model of CaV1.3 CI region docked to apoCaM. The N-lobe of apoCaM is thought to preassociate with the PCI region (green), whereas the C-lobe binds to the IQ domain (blue). The NSCaTE segment (tan) is unoccupied. This model corresponds to configuration A in Fig. 3 D, where the channels are charged with an apoCaM and ready to undergo CDI. (B) Proposed model for the Ca2+ inactivated state of the channel. The N-lobe of Ca2+/CaM is shown binding to the NSCaTE segment based on a recent NMR structure (Protein Data Bank accession no. 2LQC). This configuration is believed to result in N-lobe CDI. The de novo molecular model shows C-lobe of Ca2+/CaM, the EF hand segment, and the IQ domain forming a tripartite complex resulting in C-lobe CDI. Overall, this model corresponds to the inactive configuration ICN in Fig. 3 D. (C) Proposed allosteric mechanism of CaBP action. CaBP and apoCaM may dually bind the CaV1 channel. However, CaBP may allosterically inhibit CDI of CaV1 channels by interacting with the NT, III-IV loop, or PCI segment, thereby preventing Ca2+/CaM from reaching its effector configuration. (A–C) Adapted with permission from Yang et al. (2014).Under the regimen in Fig. 4 A, where all configurations are potentially accessible, diminished CDI by alanine substitutions occurred not only in the IQ domain, but upstream in the EF hand regions (between LGF and TLF residues). The latter effects strongly suggested that something outside the IQ domain was essential. Some of these deficits in CDI could be attributed to weakening of apoCaM preassociation with channels, because overexpressing wild-type CaM to depopulate configuration E (in Fig. 4 B) substantially recovered many of these CDI deficits. Binding assays of apoCaM with various CI segments confirmed this interpretation (Bazzazi et al., 2013; Ben Johny et al., 2013).Turning to potential deficits relating to diminished Ca2+/CaM action through channel effector sites, Fig. 4 (C and D) separately interrogates the C-lobe and N-lobe components of CDI. Importantly, isolating the C- and N-lobe components minimizes masking of mutational effects by positive cooperativity, allowing CDI deficits to be more sensitively observed in this regimen. That said, Fig. 4 C displays C-lobe CDI deficits in the EF hand region (strongest for LGF substitution), as well as in the IQ domain (with the strongest effect upon substitution at the central isoleucine). These outcomes support a hypothesis where the Ca2+/CaM effector configuration for the C-lobe component of CDI involves both of these functional hotspots, as developed two paragraphs below. For the N-lobe component of CDI (Fig. 4 D), no appreciable deficits were observed, as would be expected if interaction with the channel NSCaTE element (in the channel amino terminus) serves as the effector element.Fig. 5 A displays a proposed configuration A for apoCaM interaction with the channel. This includes a homology model of the apoCaM C-lobe complexed with the IQ domain (blue), based on analogous NaV structures (Chagot and Chazin, 2011; Feldkamp et al., 2011). The portrayal of the apoCaM N-lobe incorporates ab initio structural prediction of the CI domain, with two vestigial EF hands (EF), and a protruding helix (preIQ subelement). The EF hand module (EF) resembles the structure of a homologous NaV segment (Chagot et al., 2009; Wang et al., 2012), and the preIQ helix resembles a helical segment observed in crystal structures of analogous CaV1.2 peptides (Fallon et al., 2009; Kim et al., 2010). The atomic structure of the apoCaM N-lobe (1CFD) was interfaced with shape-complementarity docking algorithms. Regarding the proposed interaction interface of the N-lobe with the channel EF domain, we note that alanine substitution therein produced a curious enhancement of N-lobe CDI seen with certain alanine substitutions, especially in the EF region (Fig. 4 D). This feature is interesting because weakening of channel interaction with the N-lobe of apoCaM would be predicted to strengthen CDI produced by the N-lobe of Ca2+/CaM (Tadross et al., 2008).Fig. 5 B displays a proposed arrangement for the Ca2+/CaM-bound configuration ICN. The N-lobe complex with NSCaTE is an NMR structure (Liu and Vogel, 2012). An alternative ab initio model of the PCI is computationally docked with the C-lobe of Ca2+/CaM (Protein Data Bank accession no. 3BXL) and IQ module, which together form a ternary complex. This ternary arrangement is consistent with the importance of both IQ and EF domains for C-lobe CDI (Fig. 4 C). Intriguingly the C-lobe configuration resembles a rather canonical CaM–peptide complex, where the channel EF module contributes a surrogate lobe of CaM. Importantly, assays of Ca2+/CaM binding to the IQ segment alone would correlate poorly with functional effects relating to the ternary complex, as experimental studies observe (Bazzazi et al., 2013; Ben Johny et al., 2013). Overall, the proposed framework in Fig. 5 (A and B) may aid future structural biology and structure–function work. In addition, this scheme of CaM exchange within its target molecule may generalize beyond the Ca2+ channel family.

Biological consequences and prospects for new disease therapies

The biological consequences of Ca2+ regulation by CaM promise to be wide ranging and immense. In the heart, elimination of CaV1.2 CDI by means of dominant-negative CaM elicits marked prolongation of ventricular action potential duration (APD), implicating CDI as a dominant control factor in specifying APD (Alseikhan et al., 2002; Mahajan et al., 2008). As APD is one of the main determinants of electrical stability and arrhythmias in the heart, pharmacological manipulation of such regulation looms as a future antiarrhythmic strategy (Mahajan et al., 2008; Anderson and Mohler, 2009). Most recently, genome-wide linkage analysis in humans has uncovered heritable and de novo CaM mutations as the probable cause of several cases of catecholaminergic polymorphic ventricular tachycardia (CPVT), with altered CaM-ryanodine receptor function implicated as a major contributing factor (Nyegaard et al., 2012). Whole-exome sequencing has revealed an association between three de novo missense CaM mutations and severe long-QT (LQT) syndrome with recurrent cardiac arrest (Crotti et al., 2013), quite plausibly acting via disruption of CaV1.2/1.3 channel CDI (Limpitikul et al., 2014).In brain, state-of-the-art analysis of genome-wide single-nucleotide polymorphisms (SNPs) has identified CaV channels as a major risk factor for several psychiatric disorders (Cross-Disorder Group of the Psychiatric Genomics Consortium, 2013). More specifically to calmodulation, CaV1.3 channels constitute a prominent Ca2+ entry portal into pacemaking and oscillatory neurons (Bean, 2007), owing to the more negative voltages required to open these ion channels. These channels are subject to extensive alternative splicing (Hui et al., 1991; Xu and Lipscombe, 2001; Shen et al., 2006; Bock et al., 2011; Tan et al., 2011) and RNA editing (Huang et al., 2012) in the carboxy tail, in ways that strongly modulate the strength of CDI (Shen et al., 2006; Liu et al., 2010; Huang et al., 2012). This fine tuning of CDI appears to be important for circadian rhythms (Huang et al., 2012). From the specific perspective of disease, CaV1.3 channels contribute a substantial portion of Ca2+ entry into substantia nigral neurons (Bean, 2007; Chan et al., 2007; Puopolo et al., 2007; Guzman et al., 2009), which exhibit high-frequency pacemaking (Chan et al., 2007) that drives dopamine release important for movement control. Notably, loss of these neurons is intimately related to Parkinson’s disease, and Ca2+ disturbances and overload are critical to this neurodegeneration (Bezprozvanny, 2009; Surmeier and Sulzer, 2013). Accordingly, an attractive therapeutic possibility for Parkinson’s disease involves discovery of small molecules that selectively down-regulate the opening of CaV1.3 versus other closely related Ca2+ channels (Kang et al., 2012). Thus, understanding the mechanisms underlying the modulation of CaV1.3 CDI is crucial, particularly to furnish specific molecular interfaces as targets of rational screens for small-molecule modulators.The fundamental mechanism of action of a natural modulator of CaV1.3 CDI may be especially relevant. In particular, recent discoveries identify Ca2+-binding proteins (CaBPs), a family of CaM-like brain molecules (Haeseleer et al., 2000) that may also bind CaV channels and other targets (Yang et al., 2002; Kasri et al., 2004). Like CaM, CaBPs are bilobed, with each lobe containing two EF hand Ca2+ binding motifs (Haeseleer et al., 2000), and a recent crystal structure shows overall similarity to CaM (Findeisen and Minor, 2010). However, whereas all four EF hands bind Ca2+ in CaM, one lobe is nonfunctional in CaBPs. Coexpressing CaBPs with CaV1 channels eliminates their CDI, thereby potentially influencing diverse biological processes (Zhou et al., 2004; Yang et al., 2006). Some have argued that CaBPs may simply compete for apoCaM on channels (Lee et al., 2002; Findeisen et al., 2013; Oz et al., 2013). More recent data argue for a different mechanism (Fig. 5 C), in which CaBP and apoCaM can both preassociate with the channel (Yang et al., 2014). In particular, by binding to channel regions that overlap Ca2+/CaM effector loci, CaBPs may preemptively retard the ability of Ca2+/CaM to reach its effector configuration (Fig. 5 B), allowing low concentrations of CaBP molecules in the CNS to still exert functional effects in the presence of much higher CaM levels (Yang et al., 2014). If this scheme is correct, the basic mechanisms of CaBP action may have implications for drug discovery, in that small molecules that target channel interaction surfaces for CaBP and/or Ca2+/CaM may exert potent modulatory actions.In conclusion, this overview of the calmodulation of voltage-gated Ca2+ channels highlights an impressive synergy among elegant molecular regulatory mechanisms, vital biological functions, and pathogenesis and potential therapy. As such, this field promises considerable mystery, enrichment, and enlightenment in the years ahead.  相似文献   

15.
16.
The role neutrophils play in cancer is a matter of debate as both pro- and anti-tumor functions have been documented. In a recent publication in Nature, Coffelt et al. identify a new mechanism where neutrophils and T cells cooperate to generate metastasis-supporting immune suppression.Neutrophils are an important component of the innate immune system and play a critical role in fighting infections and inflammation. However, they are also propagated and mobilized by tumors and accumulate at the tumor bed and in pre-metastatic and metastatic tissues where they exert important effects. There are conflicting reports regarding the role that neutrophils play in cancer. Neutrophils have been shown to have tumoricidal properties and can kill tumor cells either by direct cytotoxicity or via antibody-dependent cell-mediated cytotoxicity (ADCC). A number of reports have also shown that neutrophils can recruit and activate T cells, inducing acquired anti-tumor immune responses. On the other hand, neutrophils were shown to possess bona fide pro-tumor properties including the promotion of tumor angiogenesis, supply of pro-tumor cytokines, stimulation of growth by neutrophil elastase, and promotion of immune evasion by immunosuppression of T cells1. Confusion about the role of neutrophils in cancer might be explained by the realization that neutrophil precursors have some plasticity2 and that multiple neutrophil subsets with differing properties likely exist within tumors, which are driven by factors in the tumor microenvironment, such as TGFβ3. It appears that early on in tumor growth, both murine and human neutrophils tend to inhibit the primary cancer growth and actually recruit and activate CD8+ T cells4,5 (Figure 1), however, as tumors become larger and the microenvironment changes, the neutrophils (along with other tumor-associated cell types, such as macrophages) begin to become immunosuppressive and inhibit T cell activity2,6,7.Open in a separate windowFigure 1The consequences of the interplay between neutrophils and lymphocytes on tumor growth and metastatic progression. Neutrophils were previously shown to possess both tumor-promoting and tumor-limiting properties. Neutrophils have the capacity to propagate cytotoxic CD8+ cells, through secretion of factors such as TNFα, cathepsin G and neutrophil elastase, thereby limiting primary tumor growth. Coffelt et al.10 identified a tumor-promoting cascade where tumor-secreted IL-1β stimulates the secretion of IL-17α from γδ T cells. Consequently, neutrophils acquire a suppressive phenotype and inhibit the propagation of cytotoxic CD8+ cells, ultimately enhancing metastatic seeding in the pre-metastatic lung.In addition to their role in influencing primary tumor growth, interesting new observations have been made about the role of neutrophils in cancer metastasis. In recent years, it has become apparent that while tumor cell-autonomous traits play a key role in the metastatic process, the normal stromal cells that surround and interact with tumor cells also play a critical part in the metastatic cascade. Again, the role of neutrophils in metastasis is unclear. We, and others, recently reported that tumor-stimulated neutrophils possess anti-metastatic activity and actively limit metastatic seeding by direct elimination of tumor cells at the pre-metastatic site8,9.In contrast to these studies, Coffelt et al.10 recently presented data to show that neutrophils could enhance metastasis in the highly aggressive KEP mouse model of metastatic breast cancer. They elegantly show that depletion of neutrophils in this model leads to a dramatic reduction in spontaneous lung metastases. They further show that the combined depletion of both neutrophils and CD8+ cells results in reversal of the metastatic phenotype, implicating CD8+ cells and neutrophils as partners in crime. Looking for the mechanism through which tumors induce this metastasis-enhancing process, the authors found that several cytokines capable of inducing IL-17α release from γδ T cells are significantly increased, and showed that IL-17α was indeed required for upregulation of G-CSF, which in turn, regulated both neutrophil mobilization and activation of the immunosuppressive neutrophil phenotype (Figure 1). Finally, the authors demonstrated that it is tumor-secreted IL-1β that stimulates the release of IL-17α, inducing the unique immune suppressive phenotype in neutrophils which acquire the ability to suppress CD8+ cytotoxic T cells and directly support metastatic spread. This complex mechanism may thus be perturbed by eliminating γδ T cells, IL-17α or neutrophils, firmly supporting the author''s conclusions.Interestingly, while this novel mechanism involving the interplay between tumor-stimulated neutrophils and two distinct T cell subsets has profound implications for metastatic spread, it apparently has no significant implications for primary tumor growth.This study raises a number of interesting issues. Are IL-1β, γδ T cells or IL-17α important in other tumors? Are these results generalizable to other mouse models and to human tumors? It is unclear why the results of this paper are so different than other reports showing that neutrophils prevent metastasis8,9. Tumor type, location, size, and the timing of interventions are all likely to be important. Regardless, this paper is a sophisticated demonstration of how tumor cells, innate immune cells and adaptive immune cells have the potential to interact in a specific tumor model. This study thus provides an interesting paradigm that should be examined in other systems.  相似文献   

17.
Although the disease-relevant microtubule-associated protein tau is known to severely inhibit kinesin-based transport in vitro, the potential mechanisms for reversing this detrimental effect to maintain healthy transport in cells remain unknown. Here we report the unambiguous upregulation of multiple-kinesin travel distance despite the presence of tau, via decreased single-kinesin velocity. Interestingly, the presence of tau also modestly reduced cargo velocity in multiple-kinesin transport, and our stochastic simulations indicate that the tau-mediated reduction in single-kinesin travel underlies this observation. Taken together, our observations highlight a nontrivial interplay between velocity and travel distance for kinesin transport, and suggest that single-kinesin velocity is a promising experimental handle for tuning the effect of tau on multiple-kinesin travel distance.Conventional kinesin is a major microtubule-based molecular motor that enables long-range transport in living cells. Although traditionally investigated in the context of single-motor experiments, two or more kinesin motors are often linked together to transport the same cargo in vivo (1–4). Understanding the control and regulation of the group function of multiple kinesins has important implications for reversing failure modes of transport in a variety of human diseases, particularly neurodegenerative diseases. Tau is a disease-relevant protein enriched in neurons (5,6). The decoration of microtubules with tau is known to strongly inhibit kinesin transport in vitro (7–9), but how kinesin-based transport is maintained in the presence of high levels of tau, particularly in healthy neurons, remains an important open question. To date, no mechanism has been directly demonstrated to reverse the inhibitory effect of tau on kinesin-based transport. Here we present a simple in vitro study that demonstrates the significant upregulation of multiple-kinesin travel distance with decreasing ATP concentration, despite the presence of tau.This investigation was motivated by our recent finding that single-kinesin velocity is a key controller for multiple-kinesin travel distance along bare microtubules (10). The active stepping of each kinesin motor is stimulated by ATP (11), and each kinesin motor remains strongly bound to the microtubule between successive steps (10,11). As demonstrated for bare microtubules (10), with decreasing ATP concentrations, each microtubule-bound kinesin experiences a decreased stepping rate per unit time and spends an increased fraction of time in the strongly bound state; additional unbound kinesins on the same cargo have more time to bind to the microtubule before cargo travel terminates. Thus, reductions in single-kinesin velocity increase the probability that at least one kinesin motor will remain bound to the microtubule per unit time, thereby increasing the travel distance of each cargo (10). Because this effect only pertains to the stepping rate of each individual kinesin and does not address the potential presence of roadblocks such as tau on the microtubules, we hypothesized in this study that single-kinesin velocity may be exploited to relieve the impact of tau on multiple-kinesin travel distance.We focused our in vitro investigation on human tau 23 (htau23, or 3RS tau), an isoform of tau that exhibits the strongest inhibitory effect on kinesin-based transport (7–9). Importantly, htau23 does not alter the stepping rate of individual kinesins (7,9), supporting our hypothesis and enabling us to decouple single-kinesin velocity from the potential effects of tau. We carried out multiple-kinesin motility experiments using polystyrene beads as in vitro cargos (8,10), ATP concentration as an in vitro handle to controllably tune single-kinesin velocity (10,11), and three input kinesin concentrations to test the generality of potential findings for multiple-kinesin transport. Combined with previous two-kinesin studies (10,12), our measurements of travel distance (Fig. 1 A) indicate that the lowest kinesin concentration employed (0.8 nM) corresponds to an average of ∼2–3 kinesins per cargo. Note that in the absence of tau, the observed decrease in bead velocity at the higher kinesin concentrations (Fig. 1 A) is consistent with a recent in vitro finding (13). At 1 mM ATP, htau23 reduced kinesin-based travel distance by a factor of two or more (Fig. 1, A and B). This observation is in good agreement with previous reports (7,8).Open in a separate windowFigure 1Distributions of multiple-kinesin travel distances measured at three experimental conditions, to verify the effect of tau (A and B) and to investigate the impact of single-kinesin velocity on the tau effect (B and C). Shaded bars at 8.7 μm indicate counts of travel exceeding the field of view. The mean travel distance (d; ± standard error of mean, SEM), sample size (n), and corresponding mean velocity (v; ± SEM) are indicated. MT denotes microtubule. Mean travel distance increased substantially at 20 μM ATP (C), despite the presence of htau23. This effect persisted across all three kinesin concentrations tested (left to right).Consistent with our hypothesis, reducing the available ATP concentration to 20 μM increased the multiple-kinesin travel distance by >1.4-fold for all three input kinesin concentrations (Fig. 1, B and C), despite the presence of htau23. The corresponding reduction in single-kinesin velocity with decreasing ATP concentration (10,11) is reflected in the ∼3.4-fold reduction in the measured bead velocities (Fig. 1, B and C). Therefore, the strong negative relationship between single-kinesin velocity and multiple-kinesin travel distance occurs not only for bare microtubules (10), but also for tau-decorated microtubules.What causes the observed increase in travel distance at the lower ATP concentration (Fig. 1, B and C)? In addition to the mechanism discussed above for the case of bare microtubules (10), an intriguing mechanism was suggested by recent studies of tau-microtubule interactions in which htau23 was observed to dynamically diffuse along microtubule lattices (14,15): reducing the stepping rate of a microtubule-bound kinesin may effectively increase the probability that a tau roadblock can diffuse away before the kinesin takes its next step.Perhaps surprisingly, although htau23 does not impact single-kinesin velocity (7,9), we observed a modest reduction in the average velocity of multiple-kinesin transport in experiments using tau-decorated microtubules (Fig. 1, A and B). This decreased velocity reflects a substantially larger variance in the instantaneous velocity for bead trajectories in the presence of htau23 (see Fig. S1 in the Supporting Material), as quantified by parsing each bead trajectory into a series of constant-velocity segments using a previously developed automatic software incorporating Bayesian statistics (16).To test the possibility that single-kinesin travel distance impacts multiple-kinesin velocity, we performed stochastic simulations (see the Supporting Material) that assumed N identical kinesin motors available for transport and included kinesin’s detachment kinetics (17). Previously, this model successfully captured multiple-dynein travel distances in vivo using single-dynein characteristics measured in vitro (18). In this study, we introduced one (and only one) free parameter to reflect the probability of each bound kinesin encountering tau at each step. When encountering tau, each kinesin has a 54% probability of detaching from the microtubule (interpolated from Fig. 2A of Dixit et al. (7)); the undetached kinesin is assumed to remain engaged in transport and completes its step along the microtubule despite the presence of tau.Remarkably, our simple simulation suggested that the tau-mediated reduction in single-kinesin travel is sufficient to reduce multiple-kinesin velocity (Fig. 2 A). The majority of the velocity decrease is predicted to occur at the transition from single-kinesin to two-kinesin transport (Fig. 2). Further decreases in cargo velocity with increasing motor number are predicted to be modest and largely independent of tau (Fig. 2 B). The results of our simulation remain qualitatively the same when evaluated at two bounds (40 and 65%) encompassing the interpolated 54% probability of kinesin detaching at tau (see Fig. S2).Open in a separate windowFigure 2Stochastic simulations predict a tau-dependent reduction in multiple-kinesin velocity, assuming that the only effect of tau protein is to prematurely detach kinesin from the microtubule (or, to reduce single-kinesin travel distance). (A) Average velocity of cargos carried by the indicated number of kinesins was evaluated at 1 mM ATP, and for four probabilities that a kinesin may encounter tau at each step. Mean velocity was evaluated using 600 simulated trajectories for all simulation conditions. Error bars indicate SEM. (B) Change in cargo velocity with each additional kinesin (ΔVel/kinesin) as a function of tau-encounter probability. These values were calculated from cargo velocities shown in panel A. Error bars indicate SEM.We note that our simple simulations do not consider the possibility that kinesin may pause in front of a tau roadblock, as previously reported in Dixit et al. (7). We omitted this consideration because the interaction strength between kinesin and the microtubule in such a paused state is unknown. In a multiple-motor geometry, could a paused kinesin be dragged along by the other motors bound to the same cargo? Could a tau roadblock be forcefully swept off the microtubule surface by the collective motion of the cargo-motor complex? Significant experimental innovations are necessary to specifically address these questions in future multiple-motor assays and to guide modeling efforts. Nonetheless, our simple simulation demonstrates that reducing single-kinesin travel distance is sufficient to decrease multiple-kinesin travel distance.Taken together, our observations highlight a nontrivial interplay between velocity and travel distance for kinesin-based transport in the presence of tau. We uncover a previously unexplored dual inhibition of tau on kinesin-transport: in addition to limiting cargo travel distance, the tau-mediated reduction in single-kinesin travel distance also leads to a modest reduction in multiple-kinesin velocity. We provide what we believe to be the first demonstration of the unambiguous upregulation of multiple-kinesin travel distance despite the presence of tau, via reducing single-kinesin velocity, suggesting a mechanism that could be harnessed for future therapeutic interventions in diseases that result from aberrant kinesin-based transport.  相似文献   

18.
Unwinding of the replication origin and loading of DNA helicases underlie the initiation of chromosomal replication. In Escherichia coli, the minimal origin oriC contains a duplex unwinding element (DUE) region and three (Left, Middle, and Right) regions that bind the initiator protein DnaA. The Left/Right regions bear a set of DnaA-binding sequences, constituting the Left/Right-DnaA subcomplexes, while the Middle region has a single DnaA-binding site, which stimulates formation of the Left/Right-DnaA subcomplexes. In addition, a DUE-flanking AT-cluster element (TATTAAAAAGAA) is located just outside of the minimal oriC region. The Left-DnaA subcomplex promotes unwinding of the flanking DUE exposing TT[A/G]T(T) sequences that then bind to the Left-DnaA subcomplex, stabilizing the unwound state required for DnaB helicase loading. However, the role of the Right-DnaA subcomplex is largely unclear. Here, we show that DUE unwinding by both the Left/Right-DnaA subcomplexes, but not the Left-DnaA subcomplex only, was stimulated by a DUE-terminal subregion flanking the AT-cluster. Consistently, we found the Right-DnaA subcomplex–bound single-stranded DUE and AT-cluster regions. In addition, the Left/Right-DnaA subcomplexes bound DnaB helicase independently. For only the Left-DnaA subcomplex, we show the AT-cluster was crucial for DnaB loading. The role of unwound DNA binding of the Right-DnaA subcomplex was further supported by in vivo data. Taken together, we propose a model in which the Right-DnaA subcomplex dynamically interacts with the unwound DUE, assisting in DUE unwinding and efficient loading of DnaB helicases, while in the absence of the Right-DnaA subcomplex, the AT-cluster assists in those processes, supporting robustness of replication initiation.

The initiation of bacterial DNA replication requires local duplex unwinding of the chromosomal replication origin oriC, which is regulated by highly ordered initiation complexes. In Escherichia coli, the initiation complex contains oriC, the ATP-bound form of the DnaA initiator protein (ATP–DnaA), and the DNA-bending protein IHF (Fig. 1, A and B), which promotes local unwinding of oriC (1, 2, 3, 4). Upon this oriC unwinding, two hexamers of DnaB helicases are bidirectionally loaded onto the resultant single-stranded (ss) region with the help of the DnaC helicase loader (Fig. 1B), leading to bidirectional chromosomal replication (5, 6, 7, 8). However, the fundamental mechanism underlying oriC-dependent bidirectional DnaB loading remains elusive.Open in a separate windowFigure 1Schematic structures of oriC, DnaA, and the initiation complexes. A, the overall structure of oriC. The minimal oriC region and the AT-cluster region are indicated. The sequence of the AT-cluster−DUE (duplex-unwinding element) region is also shown below. The DUE region (DUE; pale orange bars) contains three 13-mer repeats: L-DUE, M-DUE, and R-DUE. DnaA-binding motifs in M/R-DUE, TT(A/G)T(T), are indicated by red characters. The AT-cluster region (AT cluster; brown bars) is flanked by DUE outside of the minimal oriC. The DnaA-oligomerization region (DOR) consists of three subregions called Left-, Middle-, and Right-DOR. B, model for replication initiation. DnaA is shown as light brown (for domain I–III) and darkbrown (for domain IV) polygons (right panel). ATP–DnaA forms head-to-tail oligomers on the Left- and Right-DORs (left panel). The Middle-DOR (R2 box)-bound DnaA interacts with DnaA bound to the Left/Right-DORs using domain I, but not domain III, stimulating DnaA assembly. IHF, shown as purple hexagons, bends DNA >160° and supports DUE unwinding by the DnaA complexes. M/R-DUE regions are efficiently unwound. Unwound DUE is recruited to the Left-DnaA subcomplex and mainly binds to R1/R5M-bound DnaA molecules. The sites of ssDUE-binding B/H-motifs V211 and R245 of R1/R5M-bound DnaA molecules are indicated (pink). Two DnaB homohexamer helicases (light green) are recruited and loaded onto the ssDUE regions with the help of the DnaC helicase loader (cyan). ss, single stranded.The minimal oriC region consists of the duplex unwinding element (DUE) and the DnaA oligomerization region (DOR), which contains specific arrays of 9-mer DnaA-binding sites (DnaA boxes) with the consensus sequence TTA[T/A]NCACA (Fig. 1A) (3, 4). The DUE underlies the local unwinding and contains 13-mer AT-rich sequence repeats named L-, M-, and R-DUE (9). The M/R-DUE region includes TT[A/G]T(A) sequences with specific affinity for DnaA (10). In addition, a DUE-flanking AT-cluster (TATTAAAAAGAA) region resides just outside of the minimal oriC (Fig. 1A) (11). The DOR is divided into three subregions, the Left-, Middle-, and Right-DORs, where DnaA forms structurally distinct subcomplexes (Fig. 1A) (8, 12, 13, 14, 15, 16, 17). The Left-DOR contains high-affinity DnaA box R1, low-affinity boxes R5M, τ1−2, and I1-2, and an IHF-binding region (17, 18, 19, 20). The τ1 and IHF-binding regions partly overlap (17).In the presence of IHF, ATP–DnaA molecules cooperatively bind to R1, R5M, τ2, and I1-2 boxes in the Left-DOR, generating the Left-DnaA subcomplex (Fig. 1B) (8, 17). Along with IHF causing sharp DNA bending, the Left-DnaA subcomplex plays a leading role in DUE unwinding and subsequent DnaB loading. The Middle-DOR contains moderate-affinity DnaA box R2. Binding of DnaA to this box stimulates DnaA assembly in the Left- and Right-DORs using interaction by DnaA N-terminal domain (Fig. 1B; also see below) (8, 12, 14, 16, 21). The Right-DOR contains five boxes (C3-R4 boxes) and cooperative binding of ATP–DnaA molecules to these generates the Right-DnaA subcomplex (Fig. 1B) (12, 18). This subcomplex is not essential for DUE unwinding and plays a supportive role in DnaB loading (8, 15, 17). The Left-DnaA subcomplex interacts with DnaB helicase, and the Right-DnaA subcomplex has been suggested to play a similar role (Fig. 1B) (8, 13, 16).In the presence of ATP–DnaA, M- and R-DUE adjacent to the Left-DOR are predominant sites for in vitro DUE unwinding: unwinding of L-DUE is less efficient than unwinding of the other two (Fig. 1B) (9, 22, 23). Deletion of L-DUE or the whole DUE inhibits replication of oriC in vitro moderately or completely, respectively (23). A chromosomal oriC Δ(AT-cluster−L-DUE) mutant with an intact DOR, as well as deletion of Right-DOR, exhibits limited inhibition of replication initiation, whereas the synthetic mutant combining the two deletions exhibits severe inhibition of cell growth (24). These studies suggest that AT-cluster−L-DUE regions stimulate replication initiation in a manner concerted with Right-DOR, although the underlying mechanisms remain elusive.DnaA consists of four functional domains (Fig. 1B) (4, 25). Domain I supports weak domain I–domain I interaction and serves as a hub for interaction with various proteins such as DnaB helicase and DiaA, which stimulates ATP–DnaA assembly at oriC (26, 27, 28, 29, 30). Two or three domain I molecules of the oriC–DnaA subcomplex bind a single DnaB hexamer, forming a stable higher-order complex (7). Domain II is a flexible linker (28, 31). Domain III contains AAA+ (ATPase associated with various cellular activities) motifs essential for ATP/ADP binding, ATP hydrolysis, and DnaA–DnaA interactions in addition to specific sites for ssDUE binding and a second, weak interaction with DnaB helicase (1, 4, 8, 10, 19, 25, 32, 33, 34, 35). Domain IV bears a helix-turn-helix motif with specific affinity for the DnaA box (36).As in typical AAA+ proteins, a head-to-tail interaction underlies formation of ATP–DnaA pentamers on the DOR, where the AAA+ arginine-finger motif Arg285 recognizes ATP bound to the adjacent DnaA protomer, promoting cooperative ATP–DnaA binding (Fig. 1B) (19, 32). DnaA ssDUE-binding H/B-motifs (Val211 and Arg245) in domain III sustain stable unwinding by directly binding to the T-rich (upper) strand sequences TT[A/G]T(A) within the unwound M/R-DUE (Fig. 1B) (8, 10). Val211 residue is included in the initiator-specific motif of the AAA+ protein family (10). For DUE unwinding, ssDUE is recruited to the Left-DnaA subcomplex via DNA bending by IHF and directly interacts with H/B-motifs of DnaA assembled on Left-DOR, resulting in stable DUE unwinding competent for DnaB helicase loading; in particular, DnaA protomers bound to R1 and R5M boxes play a crucial role in the interaction with M/R-ssDUE (Fig. 1B) (8, 10, 17). Collectively, these mechanisms are termed ssDUE recruitment (4, 17, 37).Two DnaB helicases are thought to be loaded onto the upper and lower strands of the region including the AT-cluster and DUE, with the aid of interactions with DnaC and DnaA (Fig. 1B) (25, 38, 39). DnaC binding modulates the closed ring structure of DnaB hexamer into an open spiral form for entry of ssDNA (40, 41, 42, 43). Upon ssDUE loading of DnaB, DnaC is released from DnaB in a manner stimulated by interactions with ssDNA and DnaG primase (44, 45). Also, the Left- and Right-DnaA subcomplexes, which are oriented opposite to each other, could regulate bidirectional loading of DnaB helicases onto the ssDUE (Fig. 1B) (7, 8, 35). Similarly, recent works suggest that the origin complex structure is bidirectionally organized in both archaea and eukaryotes (146). In Saccharomyces cerevisiae, two origin recognition complexes containing AAA+ proteins bind to the replication origin region in opposite orientations; this, in turn, results in efficient loading of two replicative helicases, leading to head-to-head interactions in vitro (46). Consistent with this, origin recognition complex dimerization occurs in the origin region during the late M-G1 phase (47). The fundamental mechanism of bidirectional origin complexes might be widely conserved among species.In this study, we analyzed various mutants of oriC and DnaA in reconstituted systems to reveal the regulatory mechanisms underlying DUE unwinding and DnaB loading. The Right-DnaA subcomplex assisted in the unwinding of oriC, dependent upon an interaction with L-DUE, which is important for efficient loading of DnaB helicases. The AT-cluster region adjacent to the DUE promoted loading of DnaB helicase in the absence of the Right-DnaA subcomplex. Consistently, the ssDNA-binding activity of the Right-DnaA subcomplex sustained timely initiation of growing cells. These results indicate that DUE unwinding and efficient loading of DnaB helicases are sustained by concerted actions of the Left- and Right-DnaA subcomplexes. In addition, loading of DnaB helicases are sustained by multiple mechanisms that ensure robust replication initiation, although the complete mechanisms are required for precise timing of initiation during the cell cycle.  相似文献   

19.
It is well established that MDCK II cells grow in circular colonies that densify until contact inhibition takes place. Here, we show that this behavior is only typical for colonies developing on hard substrates and report a new growth phase of MDCK II cells on soft gels. At the onset, the new phase is characterized by small, three-dimensional droplets of cells attached to the substrate. When the contact area between the agglomerate and the substrate becomes sufficiently large, a very dense monolayer nucleates in the center of the colony. This monolayer, surrounded by a belt of three-dimensionally packed cells, has a well-defined structure, independent of time and cluster size, as well as a density that is twice the steady-state density found on hard substrates. To release stress in such dense packing, extrusions of viable cells take place several days after seeding. The extruded cells create second-generation clusters, as evidenced by an archipelago of aggregates found in a vicinity of mother colonies, which points to a mechanically regulated migratory behavior.Studying the growth of cell colonies is an important step in the understanding of processes involving coordinated cell behavior such as tissue development, wound healing, and cancer progression. Apart from extremely challenging in vivo studies, artificial tissue models are proven to be very useful in determining the main physical factors that affect the cooperativity of cells, simply because the conditions of growth can be very well controlled. One of the most established cell types in this field of research is the Madin-Darby canine kidney epithelial cell (MDCK), originating from the kidney distal tube (1). A great advantage of this polarized epithelial cell line is that it retained the ability for contact inhibition (2), which makes it a perfect model system for studies of epithelial morphogenesis.Organization of MDCK cells in colonies have been studied in a number of circumstances. For example, it was shown that in three-dimensional soft Matrigel, MDCK cells form a spherical enclosure of a lumen that is enfolded by one layer of polarized cells with an apical membrane exposed to the lumen side (3). These structures can be altered by introducing the hepatocyte growth factor, which induces the formation of linear tubes (4). However, the best-studied regime of growth is performed on two-dimensional surfaces where MDCK II cells form sheets and exhibit contact inhibition. Consequently, the obtained monolayers are well characterized in context of development (5), mechanical properties (6), and obstructed cell migration (7–9).Surprisingly, in the context of mechanics, several studies of monolayer formation showed that different rigidities of polydimethylsiloxane gels (5) and polyacrylamide (PA) gels (9) do not influence the nature of monolayer formation nor the attainable steady-state density. This is supposedly due to long-range forces between cells transmitted by the underlying elastic substrate (9). These results were found to agree well with earlier works on bovine aortic endothelial cells (10) and vascular smooth muscle cells (11), both reporting a lack of sensitivity of monolayers to substrate elasticity. Yet, these results are in stark contrast with single-cell experiments (12–15) that show a clear response of cell morphology, focal adhesions, and cytoskeleton organization to substrate elasticity. Furthermore, sensitivity to the presence of growth factors that are dependent on the elasticity of the substrate in two (16) and three dimensions (4) makes this result even more astonishing. Therefore, we readdress the issue of sensitivity of tissues to the elasticity of the underlying substrate and show that sufficiently soft gels induce a clearly different tissue organization.We plated MDCK II cells on soft PA gels (Young’s modulus E = 0.6 ± 0.2 kPa), harder PA gels (E = 5, 11, 20, 34 kPa), and glass, all coated with Collagen-I. Gels were prepared following the procedure described in Rehfeldt et al. (17); rigidity and homogeneity of the gels was confirmed by bulk and microrheology (see the Supporting Material for comparison). Seeding of MDCK II cells involved a highly concentrated solution dropped in the middle of a hydrated gel or glass sample. For single-cell experiments, cells were dispersed over the entire dish. Samples were periodically fixed up to Day 12, stained for nuclei and actin, and imaged with an epifluorescence microscope. Details are described in the Supporting Material.On hard substrates and glass it was found previously that the area of small clusters expands exponentially until the movement of the edge cannot keep up with the proliferation in the bulk (5). Consequently, the bulk density increases toward the steady state, whereas the density of the edge remains low. At the same time, the colony size grows subexponentially (5). This is what we denote “the classical regime of growth”. Our experiments support these observations for substrates with E ≥ 5 kPa. Specifically, on glass, colonies start as small clusters of very low density of 700 ± 200 cells/mm2 (Fig. 1, A and B), typically surrounded by a strong actin cable (Fig. 1, B and C). Interestingly, the spreading area of single cells (Fig. 1 A) on glass was found to be significantly larger, i.e., (2.0 ± 0.9) × 10−3 mm2. After Day 4 (corresponding cluster area of 600 ± 100 mm2), the density in the center of the colony reached the steady state with 6,800 ± 500 cells/mm2, whereas the mean density of the edge profile grew to 4,000 ± 500 cells/mm2. This density was retained until Day 12 (cluster area 1800 ± 100 mm2), which is in agreement with previous work (9).Open in a separate windowFigure 1Early phase of cluster growth on hard substrates. (A) Well-spread single cells, and small clusters with a visible actin cable 6 h after seeding. (B) Within one day, clusters densify and merge, making small colonies. (C) Edge of clusters from panel B.In colonies grown on 0.6 kPa gels, however, we encounter a very different growth scenario. The average spreading area of single cells is (0.34 ± 0.3) × 10−3 mm2, which is six times smaller than on glass substrates (Fig. 2 A). Clusters of only few cells show that cells have a preference for cell-cell contacts (a well-established flat contact zone can be seen at the cell-cell interface in Fig. 2 A) rather than for cell-substrate contacts (contact zone is diffusive and the shape of the cells appears curved). The same conclusion emerges from the fact that dropletlike agglomerates, resting on the substrate, form spontaneously (Fig. 2 A), and that attempts to seed one single cluster of 90,000 cells fail, resulting in a number of three-dimensional colonies (Fig. 2 A). When the contact area with the substrate exceeds 4.7 × 10−3 mm2, a monolayer appears in the center of such colonies (Fig. 2 B). The colonies can merge, and if individual colonies are small, the collapse into a single domain is associated with the formation of transient irregular structures (Fig. 2 B). Ultimately, large elliptical colonies (average major/minor axis of e = 1.8 ± 0.6) with a smooth edge are formed (Fig. 2 C), unlike on hard substrates where circular clusters (e = 1.06 ± 0.06) with a ragged edge comprise the characteristic phenotype.Open in a separate windowFigure 2Early phase of cluster growth on soft substrates. (A) Twelve hours after seeding, single cells remain mostly round and small. They are found as individual, or within small, three-dimensional structures (top). The latter nucleate a monolayer in their center (bottom), if the contact area with the substrate exceeds ∼5 × 10−3 mm2. (B) Irregularly-shaped clusters appear due to merging of smaller droplets. A stable monolayer surrounded by a three-dimensional belt of densely packed cells is clearly visible, even in larger structures. (C) All colonies are recorded on Day 4.Irrespective of cluster size, in the new regime of growth, the internal structure is built of two compartments (Fig. 2 B):
  • 1.The first is the edge (0.019 ± 0.05-mm wide), a three-dimensional structure of densely packed cells. This belt is a signature of the new regime because on hard substrates the edge is strictly two-dimensional (Fig. 1 C).
  • 2.The other is the centrally placed monolayer with a spatially constant density that is very weakly dependent on cluster size and age (Fig. 3). The mean monolayer density is 13,000 ± 2,000 cells/mm2, which is an average over 130 clusters that are up to 12 days old and have a size in the range of 10−3 to 10 mm2, each shown by a data point in Fig. 3. This density is twice the steady-state density of the bulk tissue in the classical regime of growth.Open in a separate windowFigure 3Monolayer densities in colonies grown on 0.6 kPa substrates, as a function of the cluster size and age. Each cluster is represented by a single data point signifying its mean monolayer density. (Black lines) Bulk and (red dashed lines) edge of steady-state densities from monolayers grown on glass substrates. Error bars are omitted for clarity, but are discussed in the Supporting Material.
Until Day 4, the monolayer is very homogeneous, showing a nearly hexagonal arrangement of cells. From Day 4, however, defects start to appear in the form of small holes (typical size of (0.3 ± 0.1) × 10−3 mm2). These could be attributed to the extrusions of viable cells, from either the belt or areas of increased local density in the monolayer (inset in Fig. 4). This suggests that extrusions serve to release stress built in the tissue, and, as a consequence, the overall density is decreased.Open in a separate windowFigure 4Cell nuclei within the mother colony and in the neighboring archipelago of second-generation clusters grown on 0.6 kPa gels at Day 12. (Inset; scale bar = 10 μm) Scar in the tissue, a result of a cell-extrusion event. (Main image; scale bar = 100 μm) From the image of cell nuclei (left), it is clear that there are no cells within the scar, whereas the image of actin (right) shows that the cytoplasm of the cells at the edge has closed the hole.Previous reports suggest that isolated MDCK cells undergo anoikis 8 h after losing contact with their neighbors (18). However, in this case, it appears that instead of dying, the extruded cells create new colonies, which can be seen as an archipelago surrounding the mother cluster (Fig. 4). The viability of off-cast cells is further evidenced by the appearance of single cells and second-generation colonies with sizes varying over five orders of magnitude, from Day 4 until the end of the experiment, Day 12. Importantly, no morphological differences were found in the first- and second-generation colonies.In conclusion, we show what we believe to be a novel phase of growth of MDCK model tissue on soft PA gels (E = 0.6 kPa) that, to our knowledge, despite previous similar efforts (9), has not been observed before. This finding is especially interesting in the context of elasticity of real kidneys, for which a Young’s modulus has been found to be between 0.05 and 5 kPa (19,20). This coincides with the elasticity of substrates studied herein, and opens the possibility that the newly found phase of growth has a particular biological relevance. Likewise, the ability to extrude viable cells may point to a new migratory pathway regulated mechanically by the stresses in the tissue, the implication of which we hope to investigate in the future.  相似文献   

20.
Maintenance of hematopoietic stem cells (HSCs) in vitro has been believed to be difficult due to a lack of complete understanding of HSC quiescence maintained by the niche. Recent evidence suggests that in vitro maintenance of human and mouse long-term HSCs (LT-HSCs) is possible through dual inhibition (2i) of both GSK-3 and mTOR in the absence of cytokines, serum, or feeder cells.Hematopoietic stem cells (HSCs) are generally quiescent, and have the ability to self-renew or to differentiate into mature blood cells. Despite recent advances, it has not been possible to maintain functional long-term HSCs (LT-HSCs) outside the hematopoietic niche, because mechanisms by which HSC quiescence is maintained by the niche1 have not been fully understood. There have been many attempts to expand HSCs and hematopoietic progenitor cells in vitro using hematopoietic cytokines combined with factors, including Wnt activators2,3,4, glycogen synthase kinase 3 (GSK-3) inhibitors5, Notch ligand, HoxB4, prostaglandin E2, aryl hydrocarbon receptor antagonists, angiopoietin-like proteins, or pleiotrophin6,7. However, all studies have required hematopoietic cytokines, which may promote lineage commitment at the expense of LT-HSC maintenance.Huang et al.8 previously reported that disruption of GSK-3 in hematopoietic cells in mice leads to an increase in the number of HSCs through Wnt activation, and that the subsequent depletion of LT-HSCs occurs because inhibition of GSK-3 also activates mammalian target of rapamycin (mTOR) (Figure 1A). The mTOR pathway is recognized as an established nutrient sensor, and nutrient-sensing systems are associated with HSC homeostasis. Indeed, HSCs reside in a low-perfusion environment in the bone marrow with low oxygen and low nutrition. Activation of mTOR has been shown to increase the proliferation of committed progenitors at the cost of HSC maintenance (Figure 1A), indicating that low nutrient availability is an essential characteristic of the niche. Thus, Huang et al. hypothesized that low nutrient availability might contribute to HSC maintenance.Open in a separate windowFigure 1Schematic diagrams of maintenance of LT-HSCs. (A) Disruption of GSK-3 results in HSC self-renewal through Wnt activation in a β-catenin-dependent manner. However, in assays of LT-HSC function, disruption of GSK-3 leads to depletion of HSCs through activation of mTOR. GSK-3 regulates both Wnt and mTOR signalings in HSCs8. (B) GSK-3 inhibition-induced mTOR activation is attributable to HSC depletion, which can be prevented by mTOR inhibition9. (C) In vitro maintenance of LT-HSCs is possible by dual inhibition (2i) of GSK-3 and mTOR under cytokine-free, serum-free, feeder-free conditions9. CHIR99021 and lithium are GSK-3 inhibitors, and rapamycin is mTOR inhibitor.Recently, Huang et al.9 clearly demonstrate that human and mouse LT-HSCs can be maintained in vitro by inhibiting both GSK-3 and mTOR, in the absence of cytokines, serum, or feeder cells (Figure 1B). Moreover, the combination of two clinically approved inhibitors, lithium (GSK-3) and rapamycin (mTOR) (Figure 1C), increases the number of functional LT-HSCs in mice. First, Huang et al.9 determined whether dual inhibition (2i) of GSK-3 and mTOR would be sufficient for maintaining HSCs in vitro. They cultured mouse c-Kit+ or LinSca1+c-Kit+ (LSK) cells in X-VIVO 15 (Lonza) (which is chemically defined, serum-free, hematopoietic cell medium) supplemented with inhibitors of GSK-3 (CHIR99021 or lithium) and mTOR (rapamycin) for 7 days in the absence of cytokines, serum, or feeder cells. They subsequently assessed the hematopoietic potential of the cultured HSCs by competitive repopulation assay. It was confirmed that HSCs cultured with 2i maintained long-term reconstitution potential, and that the frequency of HSCs was similar to that in uncultured c-Kit+ cells. Similarly, they confirmed that the effects of 2i on LT-HSCs were recapitulated in human HSCs that are present in umbilical cord blood CD34+ cells. To explore the mechanism by which 2i preserves HSCs, they also investigated cell cycle status in mouse LSK cells. They found an increased percentage of quiescent cells by 2i, suggesting that the maintenance of LSK cells by 2i is the result of increased dormancy in vitro. Finally, they demonstrated that GSK-3 and mTOR inhibition increases mouse LT-HSCs in vivo. They treated mice with lithium and rapamycin for 2 weeks, and found that both the overall bone marrow cellularity and the absolute number of LT-HSCs increased in the treatment group. In a competitive repopulation assay, the absolute number of competitive rescue units was increased by 2-fold in bone marrow of treated mice.The above findings by Huang et al.9 are outstanding, but many questions need to be answered in future studies. i) The authors examined 2i cultures for 7 days in vitro (and 2 weeks in vivo), and it would be interesting to examine how long it is possible to maintain LT-HSCs in vitro under 2i condition. However, as the authors mentioned9, prolonged activation of Wnt signaling might be associated with transformation in vitro, and might have the risk of inducing colorectal cancers and leukemias when GSK-3 inhibitors are administered in vivo10. Nevertheless, lithium (GSK-3 inhibitor) has been used to treat bipolar disorder for over 50 years and is not associated with an increased risk of malignancies11, as the authors pointed out9. ii) In cytokine-free medium, is there cytokine production by HSCs or progenitor cells? It may be possible that cytokine production would contribute to the maintenance of LT-HSCs in an autocrine or paracrine manner. iii) Although feeder cells and/or serum are not defined factors for culture, it would be of interest to investigate whether 2i culture in the presence of supporting cells would further improve the maintenance of LT-HSCs. Some extrinsic regulators for HSC quiescence12, such as N-cadherins, could contribute to LT-HSC maintenance in cooperation with 2i. iv) In addition to iii), hypoxic environment is known to be an extrinsic regulator for HSC quiescence12, as the bone marrow niche is a low-perfusion environment. Hypoxic culture might synergize with 2i. v) When human ESCs/iPSCs are induced to differentiate into HSCs, it is difficult to capture true human LT-HSCs in vitro. If this is due to inability to maintain human LT-HSCs in vitro, it would be interesting to examine whether 2i culture would enable in vitro induction and maintenance of transplantable LT-HSCs derived from human ESCs/iPSCs.In summary, dual inhibition (2i) of GSK-3 and mTOR allows for the maintenance of human and mouse LT-HSCs in vitro (Figure 1C), and this may resolve the difficulty in culturing HSCs, which in turn, may improve basic research of HSCs (e.g., gene editing in vitro) and human HSC transplantation outcomes. Furthermore, although the effect of 2i on expansion of HSCs is relatively small, a combination of 2i drugs may increase human clinical trials1,6,7 that use 2i in vivo for the aim of increasing the number of LT-HSCs, since 2i drugs are known as clinically tolerated medications. Insights gained from the discovery of 2i for HSC maintenance may lead to great benefits for patients with hematologic disorders, hopefully in the near future.  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