首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
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.
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   

3.
Teleost fishes are the most species-rich clade of vertebrates and feature an overwhelming diversity of sex-determining mechanisms, classically grouped into environmental and genetic systems. Here, we review the recent findings in the field of sex determination in fish. In the past few years, several new master regulators of sex determination and other factors involved in sexual development have been discovered in teleosts. These data point toward a greater genetic plasticity in generating the male and female sex than previously appreciated and implicate novel gene pathways in the initial regulation of the sexual fate. Overall, it seems that sex determination in fish does not resort to a single genetic cascade but is rather regulated along a continuum of environmental and heritable factors.IN contrast to mammals and birds, cold-blooded vertebrates, and among them teleost fishes in particular, show a variety of strategies for sexual reproduction (Figure 1), ranging from unisexuality (all-female species) to hermaphroditism (sequential, serial, and simultaneous, including outcrossing and selfing species) to gonochorism (two separate sexes at all life stages). The underlying phenotypes are regulated by a variety of sex determination (SD) mechanisms that have classically been divided into two main categories: genetic sex determination (GSD) and environmental sex determination (ESD) (Figure 2).Open in a separate windowFigure 1Reproductive strategies in fish. Fish can be grouped according to their reproductive strategy into unisexuals, hermaphrodites, and gonochorists. Further subdivisions of these three categories are shown with pictures of species exemplifying the strategies. Fish images: Amphiprion clarkii courtesy of Sara Mae Stieb; Hypoplectrus nigricans courtesy of Oscar Puebla; Scarus ferrugineus courtesy of Moritz Muschick; Astatotilapia burtoni courtesy of Anya Theis; Poecilia formosa and Kryptolebias marmoratus courtesy of Manfred Schartl; Trimma sp. courtesy of Rick Winterbottom [serial hermaphroditism has been described in several species of the genus Trimma (Kuwamura and Nakashima 1998; Sakurai et al. 2009; and references therein)].Open in a separate windowFigure 2Sex-determining mechanisms in fish. Sex-determining systems in fish have been broadly classified into environmental and genetic sex determination. For both classes, the currently described subsystems are shown.Environmental factors impacting sex determination in fish are water pH, oxygen concentration, growth rate, density, social state, and, most commonly, temperature (for a detailed review on ESD see, e.g., Baroiller et al. 2009b and Stelkens and Wedekind 2010). As indicated in Figure 2, GSD systems in fish compose a variety of different mechanisms and have been reviewed in detail elsewhere (e.g., Devlin and Nagahama 2002; Volff et al. 2007).The GSD systems that have received the most scientific attention so far are those involving sex chromosomes, which either may be distinguishable cytologically (heteromorphic) or appear identical (homomorphic). In both cases, one sex is heterogametic (possessing two different sex chromosomes and hence producing two types of gametes) and the other one homogametic (a genotype with two copies of the same sex chromosome, producing only one type of gamete). A male-heterogametic system is called an XX-XY system, and female-heterogametic systems are denoted as ZZ-ZW. Both types of heterogamety exist in teleosts and are even found side by side in closely related species [e.g., tilapias (Cnaani et al. 2008), ricefishes (Takehana et al. 2008), or sticklebacks (Ross et al. 2009)]; for more details on the phylogenetic distribution of GSD mechanisms in teleost fish, see Mank et al. (2006). Note that sex chromosomes in fish are mostly homomorphic and not differentiated (Ohno 1974), which is in contrast to the degenerated Y and W chromosomes in mammals (Graves 2006) and birds (Takagi and Sasaki 1974), respectively. This is one possible explanation for the viable combination of different sex chromosomal systems within a single species or population of fish (Parnell and Streelman 2013) and could be a mechanistic reason why sex chromosome turnovers occur easily and frequently in this group (Mank and Avise 2009). Additionally, fish can have more complex sex chromosomal systems involving more than one chromosome pair (see Figure 2). Even within a single fish species, more than two sex chromosomes may occur at the same time, or more than two types of sex chromosomes may co-exist in the same species (Schultheis et al. 2006; Cioffi et al. 2013), which can sometimes be due to chromosome fusions (Kitano and Peichel 2012).Detailed insights on the gene level for GSD/sex chromosomal systems are currently available for only a limited number of fish species, and all but one of these cases involve a rather simple genetic system with male heterogamety and one major sex determiner (see below). The only exception is the widely used model species zebrafish (Danio rerio), which has a polyfactorial SD system implicating four different chromosomes (chromosomes 3, 4, 5, and 16) (Bradley et al. 2011; Anderson et al. 2012) and also environmental cues (Shang et al. 2006).In this review, we focus on newly described genetic sex-determining systems and possible mechanisms allowing their emergence in fishes, which are the most successful group of vertebrates with ∼30,000 species.  相似文献   

4.
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   

5.
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   

6.
7.
8.
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   

9.
10.
11.
12.
We combine total internal reflection fluorescence structured illumination microscopy with spatiotemporal image correlation spectroscopy to quantify the flow velocities and directionality of filamentous-actin at the T cell immunological synapse. These techniques demonstrate it is possible to image retrograde flow of filamentous-actin at superresolution and provide flow quantification in the form of velocity histograms and flow vector maps. The flow was found to be retrograde and radially directed throughout the periphery of T-cells during synapse formation.Many biological processes are now being visualized with the use of superresolution fluorescence microscopy techniques. However, localization-based techniques primarily rely on fixed or slow moving samples to permit the collection of structural information. The 10-fold gains in resolution afforded by these superresolution techniques are usually possible through sacrificing the factors that originally made microscopy such a powerful tool: the ability to image live cells. In the case of stimulated emission depletion imaging, the scanning approach associated with this technique may fail to detect faster molecular events when imaging whole cellular regions.Structured illumination microscopy (SIM) is an alternative to these methods (1). It increases the resolution of conventional fluorescence microscopy twofold; it has the advantage of using a wide-field system, providing fast acquisition speeds of whole cells with relatively low laser powers; and it is compatible with standard fluorophores. By using a physical grating to produce interference patterns from a laser, periodic illumination is created. This patterned illumination causes information from higher spatial frequencies to be downmodulated (i.e., shifted) into the optical transfer function (support region) of the lens, resulting in higher-resolution spatial information being captured than is ordinarily obtainable.To quantify the directional motion of intracellular molecules, spatiotemporal image correlation spectroscopy (STICS (2)) was applied. Using spatial image correlation in time, STICS measures the similarity of pixels with those surrounding in lagging frames via a correlation function. The correlation function provides information on both flow velocities and directionality, while discounting static structures through the immobile object filter, achieved by subtracting a moving average of pixel intensities.The formation of an immunological synapse between T cells and antigen-presenting cells is a process requiring many dynamic (3) and subdiffraction-limited clustering events (4–6) to take place. The polymerization of actin is important for the spreading of cells over their target antigen-presenting cells (7), as well as cell mobility and migration (8). Retrograde flow of densely meshed cortical actin is observed at the basal membrane of synapse-forming T cells, where it may have a role in the corralling and clustering of signaling molecules at the plasma membrane (9), as well as at the leading edge of migrating cells (10). Filamentous actin is an extremely dynamic (7), densely packed, and thin (7-nm) structure (11,12).Here, we perform STICS on SIM data acquired on a total internal reflection fluorescence (TIRF) microscope system, which generated an evanescent field of 75-nm depth for excitation. To our knowledge, this is the first demonstration of an image correlation approach to quantify molecular dynamics on subresolution length scales using wide-field microscopy. To demonstrate the technique, we analyze two-dimensional actin flows in CD4+ T cells during immunological synapse formation, performed after cross-linking of antigen T cell receptors on a coverslip coated with specific antibodies.Fig. 1 a shows a schematic of the TIRF SIM setup. Excitation light (488 nm) passes through a polarizing module and then a phase-grating block, producing diffracted beams. These are then passed through a diffraction filter module to isolate the −1 and +1 order laser beams. These first-order laser beams are angled through the objective to produce total internal reflection conditions at the glass-water interface. The two evanescent waves interfere at the sample, producing structured illumination. The setup then produces lateral and rotational shifts through three orientations, producing nine raw images containing higher spatial frequencies than can normally be acquired by an objective using standard light microscopy. Fig. 1 b demonstrates the increased resolution obtained from TIRF SIM. Shown are the collected Fourier frequencies compared to those of a conventional microscope (dotted red line). Resolution was also measured using sparse 100-nm diameter fluorescent beads. Fig. 1 c shows a magnified image of these beads from which a line profile was obtained (yellow arrow). The full width at half-maximum of this profile (Fig. 1 d) gives a lateral resolution for the system of 120 nm.Open in a separate windowFigure 1(a) Schematic of the TIRF SIM setup. (b) Demonstration of the doubling of spatial resolution of collected frequencies through a Fourier transform (superimposed red circle demonstrating regular spatial frequency limits). (c) SIM reconstructed image of 100-nm bead (scale bar 0.5 μm). (d) (Plotted line) Bead showing full width at half-maximum of 120 nm.We then applied STICS analysis to quantify actin flow in T cell synapses acquired using TIRF SIM (Fig. 2). Fig. 2 a shows a schematic of the STICS analysis. From the raw data, immobile objects are first filtered by subtracting a moving average of the pixel values. Vector maps were obtained from correlation analysis of the time-series as previously published in Hebert et al. (2) and Brown et al. (13). Fig. 2 b shows a reconstructed TIRF SIM image of a mature T cell immunological synapse, representative of a time-point derived from the time series acquired at 1.28 fps (see Movie S1 in the Supporting Material). From this reconstructed image, two representative regions have been selected. In these regions, pseudo-colored actin flow vectors are overlaid onto the fluorescence intensity image. These range in magnitude from 0.01 μm/min (blue) to 5.61 μm/min (red). It can be observed that all flow vectors are directed radially toward the synapse center. A histogram of this flow is shown in Fig. 2 c. The histogram shows a peak retrograde flow velocity of 1.91 ± 1.27 μm/min. These data are representative of n = 7 T-cell synapses imaged by TIRF SIM.Open in a separate windowFigure 2(a) STICS analysis, performed by isolating mobile from immobile structures through a moving average filter (i) and binning a subset of pixels into blocks of superpixels (ii); the STICS software correlates spatial fluorescence fluctuations through time (iii). The code then outputs vector maps showing directionality and flow velocities. (b) TIRF SIM image of actin flow in a T cell 5 min after contact with a stimulatory coverslip. (Zoomed regions) Retrograde actin flow at the synapse periphery. (c) Histograms showing flow speed statistics of vectors from T-cell synapses (n = 7).  相似文献   

13.
Thomas D. Fox 《Genetics》2012,192(4):1203-1234
The mitochondrion is arguably the most complex organelle in the budding yeast cell cytoplasm. It is essential for viability as well as respiratory growth. Its innermost aqueous compartment, the matrix, is bounded by the highly structured inner membrane, which in turn is bounded by the intermembrane space and the outer membrane. Approximately 1000 proteins are present in these organelles, of which eight major constituents are coded and synthesized in the matrix. The import of mitochondrial proteins synthesized in the cytoplasm, and their direction to the correct soluble compartments, correct membranes, and correct membrane surfaces/topologies, involves multiple pathways and macromolecular machines. The targeting of some, but not all, cytoplasmically synthesized mitochondrial proteins begins with translation of messenger RNAs localized to the organelle. Most proteins then pass through the translocase of the outer membrane to the intermembrane space, where divergent pathways sort them to the outer membrane, inner membrane, and matrix or trap them in the intermembrane space. Roughly 25% of mitochondrial proteins participate in maintenance or expression of the organellar genome at the inner surface of the inner membrane, providing 7 membrane proteins whose synthesis nucleates the assembly of three respiratory complexes.TO think about how mitochondrial proteins are synthesized, imported, and assembled, it is useful to have a clear picture of the organellar structures that they, along with membrane lipids, compose and the functions that they carry out. As almost every schoolchild learns, mitochondria carry out oxidative phosphorylation, the controlled burning of nutrients coupled to ATP synthesis. Since Saccharomyces cerevisiae prefers to ferment sugars, respiration is a dispensable function and nonrespiring mutants are viable [although they cannot undergo meiosis (Jambhekar and Amon 2008)]. However, mitochondria themselves are not dispensable. A substantial fraction of intermediary metabolism occurs in mitochondria (Strathern et al. 1982), and at least one of these pathways, iron–sulfur cluster assembly, is essential for growth (Kispal et al. 2005). Thus, any mutation that prevents the biogenesis of mitochondria by, for example, preventing the import of protein constituents from the cytoplasm, is lethal (Baker and Schatz 1991).The mitochondria of S. cerevisiae are tubular structures at the cell cortex. While the number of distinct compartments can range from 1 to ∼50 depending upon conditions (Stevens 1981; Pon and Schatz 1991), continual fusion and fission events among them effectively form a single dynamic network (Nunnari et al. 1997). The outer membrane surrounds the tubules. The inner membrane has a boundary domain closely juxtaposed beneath the outer membrane and cristae domains that project internally from the boundary into the matrix (Figure 1A). The matrix is the aqueous compartment surrounded by the inner membrane. The aqueous intermembrane space lies between the membranes and is continuous with the space within cristae.Open in a separate windowFigure 1 Overview of mitochondrial structure in yeast. (A) Schematic of compartments comprising mitochondrial tubules. The outer membrane surrounds the organelle. The inner membrane surrounds the matrix and consists of two domains, the inner boundary membrane and the cristae membranes, which are joined at cristae junctions. The intermembrane space lies between the outer membrane and inner membrane. (B) Electron tomograph image of a highly contracted yeast mitochondrion observed en face (a) with the outer membrane (red) and (b) without the outer membrane. Reprinted by permission from John Wiley & Sons from Mannella et al. (2001).Inner membrane cristae are often depicted as baffles emanating from the boundary domain. However, electron tomography of mitochondria from several species, including yeast, shows that cristae actually emanate from the boundary membrane as narrow tubular structures at sites termed “crista junctions” and expand as they project into the matrix (Frey and Mannella 2000; Mannella et al. 2001) (Figure 1B). It seems clear that the boundary and cristae domains of the inner membrane have distinct compositions with respect to the respiratory complexes that are embedded preferentially in the cristae membrane domains, as well as other components (Vogel et al. 2006; Wurm and Jakobs 2006; Rabl et al. 2009; Suppanz et al. 2009; Zick et al. 2009; Davies et al. 2011).The outer and inner boundary membranes are connected at multiple contact sites, at least some of which are involved in protein translocation and may be transient (Pon and Schatz 1991). In addition, there appear to be firm contact sites, not directly involved with protein translocation, preferentially colocalized with crista junctions (Harner et al. 2011a).Overall, there appear to be ∼1000 distinct proteins in yeast mitochondria (Premsler et al. 2009). One series of proteomic studies on highly purified organelles identified 851 proteins thought to represent 85% of the total number of species (Sickmann et al. 2003; Reinders et al. 2006; Zahedi et al. 2006). Another study identified an additional 209 candidates (Prokisch et al. 2004). A computationally driven search for candidates involved in yeast mitochondrial function, coupled with experiments to assay respiratory function and maintenance of mitochondrial DNA (mtDNA), identified 109 novel candidates, although many of these may not be mitochondrial per se (Hess et al. 2009). Taking the boundary and cristae domains together, the inner membrane is the most protein-rich mitochondrial compartment, followed by the matrix (Daum et al. 1982).Only eight of the yeast mitochondrial proteins detected in proteomic studies are encoded by mtDNA and synthesized within the organelle. They are hydrophobic subunits of respiratory complexes III (bc1 complex or ubiquinol-cytochrome c reductase), IV (cytochrome c oxidase), and V (ATP synthase), as well as a hydrophilic mitochondrial small subunit ribosomal protein. The remaining ∼99% of yeast mitochondrial proteins are encoded by nuclear genes, synthesized in cytoplasmic ribosomes, and imported into the organelle.An overview of known nuclearly encoded mitochondrial protein functions (Figure 2) reveals that ∼25% of them are involved directly in genome maintenance and expression of the eight major mitochondrial genes (Schmidt et al. 2010). The functions of ∼20% of the proteins are not known. Fifteen percent are involved in the well-known processes of energy metabolism. Protein translocation, folding, and turnover functions occupy ∼10% of mitochondrial proteins.Open in a separate windowFigure 2 Classification of identified mitochondrial proteins according to function. Reprinted by permission from Nature Publishing Group from Schmidt et al. (2010).The following discussion reviews our understanding of the biogenesis of mitochondria starting on the outside, the cytoplasm, and working inward through the mitochondrial compartments.  相似文献   

14.
15.
Confocal Raman microspectroscopy and fluorescence imaging are two well-established methods providing functional insight into the extracellular matrix and into living cells and tissues, respectively, down to single molecule detection. In living tissues, however, cells and extracellular matrix coexist and interact. To acquire information on this cell-matrix interaction, we developed a technique for colocalized, correlative multispectral tissue analysis by implementing high-sensitivity, wide-field fluorescence imaging on a confocal Raman microscope. As a proof of principle, we study early stages of bone formation in the zebrafish (Danio rerio) larvae because the zebrafish has emerged as a model organism to study vertebrate development. The newly formed bones were stained using a calcium fluorescent marker and the maturation process was imaged and chemically characterized in vivo. Results obtained from early stages of mineral deposition in the zebrafish fin bone unequivocally show the presence of hydrogen phosphate containing mineral phases in addition to the carbonated apatite mineral. The approach developed here opens significant opportunities in molecular imaging of metabolic activities, intracellular sensing, and trafficking as well as in vivo exploration of cell-tissue interfaces under (patho-)physiological conditions.Understanding fundamental biological processes relies on probing intra- and extracellular environments, targeted delivery inside living cells and tissues, and real-time detection and imaging of chemical markers and biomolecules (1,2). Typically, information about molecules in cellular environments is obtained by fluorescence microscopy (3). This is a powerful imaging tool for localizing and imaging samples but requires fluorescent labels and markers and lacks capabilities for quantitative mapping of the chemical composition in complex systems. In this regard, confocal Raman spectroscopic imaging is becoming increasingly popular for label-free chemical detection, due to the inherent scattering nature of all biomolecules (4,5). However, confocal Raman imaging alone does not allow live, high-resolution imaging of larger regions of interest in complex biological tissues. Transcutaneous Raman spectroscopy has the potential as a tool for in vivo bone quality assessment (6), whereas the time- and space-resolved Raman spectroscopy allows the visualization in vivo of the distributions of molecular species in human and yeast cells (4,5,7). Here we developed a correlative Raman and fluorescence imaging method that combines the strengths and compensates for the shortcomings of each of these imaging modalities and allows studying in vivo processes in complex animal models such as zebrafish larvae. There are two main advantages of this approach over previous studies (8,9): low light intensity and high acquisition rate, making it well suited for real-time investigation of live samples.Fig. 1, a and b, shows a schematic representation of the experimental setup and of the optical path, respectively. The two techniques are implemented on a commercially available Raman microscope body to perform simultaneously confocal Raman spectroscopy and wide-field fluorescence imaging (see the Supporting Material for details of components). Briefly, the multimodality of the setup is provided by a combination of dichroic mirrors (DM 1–3) and filters that at turns reflect or transmit the excitation and emission signals. This combination of optics allows simultaneous collection of fluorescence images (2560 × 2160 pixels at 30 fps) with excitation at 400 and 490 nm and spatially resolved Raman spectra with excitation at 633 nm.Open in a separate windowFigure 1Fluorescence imaging of zebrafish larvae. (a) Cartoon of the experimental setup showing how the different modules are assembled onto the microscope for the simultaneous use of confocal Raman spectroscopy and fluorescence imaging. (b) Schematic representation of the optical path. (c) Fluorescence image of calcium-containing tissues, and fluids stained with calcein blue and excited at 400 nm (top). Endothelial cells of transgenic tg(fli1:EGFP)y1 zebrafish excited at 490 nm (bottom).As a proof of principle, we have studied the different mineral phases involved in bone formation of the zebrafish larvae. The bone development process involves the transport of ions to specific cells (osteoblasts) that are responsible for the subsequent mineral formation and deposition. The mineral phase in these cells is a poorly characterized disordered calcium phosphate (10–12). The mineral-bearing intracellular vesicles release their content into the extracellular collagen fibrils, where the mineral subsequently crystallizes as carbonated hydroxyapatite (13). Very little is known about the phase transformations the mineral undergoes after the deposition into the collagen matrix in vivo. Raman spectroscopy studies of bone tissue in organ cultures evidenced that the inorganic mineral deposition proceeds through transient intermediates including octacalcium phosphate-like (OCP) minerals (14).To assess the feasibility of imaging a vertebrate organism, fluorescence images of an entire zebrafish larva (Fig. 1 c) were acquired with the correlative fluorescence-Raman setup. The two images in Fig. 1 c were composed by merging several low-magnification (10×) fluorescence images. Larvae of transgenic zebrafish Tg(fli:EGFP); nac mutants (albino fish) expressing EGFP in the cytoplasm of endothelial cells was used. The newly formed bones were stained by soaking the live embryo noninvasively in the calcium markers calcein blue 0.2% wt or calcein green 0.2% wt.The calcein blue marker is excited at 400 nm. It is labeling bones and can be also detected as a fluorescent marker not associated with formed bones (e.g., stomach) (Fig. 1 c, top). At 490 nm, calcein green and endothelial cells within blood vessels expressing EGFP are excited (Fig. 1 c, bottom). Because EGFP and calcein blue have significantly different excitation and emissions spectra, dual staining with calcein blue (as a mineral marker) and EGFP allows fast-switching dual-wavelength fluorescence imaging. Furthermore, because the spectra of the calcium markers and EGFP do not extend beyond the Raman laser, these fluorophores are appropriate candidates for experiments requiring Raman and fluorescence imaging. The dual-excitation offers the capability of mapping several tissues in a single experiment at the video rate. This, in principle, could be used to probe different parameters of the microenvironment (e.g., pH (15), temperature (16), viscosity (17), and calcium concentration (18)) using wavelength-ratiometric fluorescence imaging which, in correlation with confocal Raman spectroscopy, could open new strategies in studies of the microenvironmental properties in living tissues.The fin rays of zebrafish are a simple, growing bone-model system, in which the fins are gradually mineralized within spatially resolved regions (19). Raman spectroscopy revealed details of the calcein green-stained fin where new bone is deposited (Fig. 2). In Fig. 2 a, a fluorescence image of a zebrafish larva analogous to the top image in Fig. 1 c is shown. The right inset in Fig. 3 b shows higher-magnification (60× water-immersion objective) details of the calcein green-stained fin typical of newly deposited bone. Raman spectra of progressively mineralized bone tissue were acquired within representative regions (Fig. 2 b; numbered 1–4). The spectra exhibit characteristic bands that can be assigned to the organic protein extracellular matrix (amide I, amide III, Phe, C-H, etc.) and the inorganic mineral content (v1, v2, v4 of PO43−).Open in a separate windowFigure 2Correlative fluorescence-Raman imaging of zebrafish fin bone maturation. (a) Low-resolution (10×) fluorescence image of zebrafish stained with calcein green, with high-resolution (60×) details (right inset in panel b) of a representative fin ray region where Raman spectra (b) of progressively mineralized bone tissue were acquired (numbered 1–4). (Left inset in panel b) Integral of the orientation independent mineral band (v2) where a clear drop of the mineral content can be observed.The analyses of the orientation-independent v2 phosphate band revealed a clear drop in the mineral content based on the intensity integral (left inset in Fig. 2 b). Assuming that the spectrum collected in region 4 contains only organic matrix (very small phosphate-related peaks) and by subtracting it from the spectrum of mineral-rich bone region (spectrum 1, proximal part of the tail bone), spectral features of only the mineral phase can be plotted (black line). In addition to the phosphate (PO43−) and carbonate (CO32−) bands assignable to the carbonated apatite phase characteristic of the more mature bone mineral, several peaks related to the hydrogen phosphate (HPO42−) species can be clearly distinguished.The HPO42− peaks are characteristic of the OCP mineral phase that has been postulated, together with amorphous calcium phosphate, as an intermediate mineral phase in the process of bone maturation (10,13,14,20), but never observed directly in living animals. Our findings show in vivo potential of the correlative setup envisioned by Crane et al. (14) and confirm that the mineral maturation indeed proceeds through an OCP-like mineral phase. Further analysis of the mineral spectrum in Fig. 2 b reveals an extremely broad band in the region 800–1100 cm−1. This envelope can be related to hydrogenated phosphate species typical of amorphous calcium phosphate precipitated in an acidic environment (see Fig. S1 in the Supporting Material), suggesting that this phase is also contributing to the maturation process.In conclusion, the methodology developed here allows for unprecedented chemical characterization of fluorescently-labeled biological tissues in vivo. The approach is suitable for long-term in vivo characterization of zebrafish bone mineralization under (patho-)physiological conditions. Furthermore, the setup can be upgraded to host other advance fluorescence imaging techniques such as super-resolution microscopy (e.g., photoactivated localization microscopy), two-photon excitation, and Forster resonance energy transfer or fluorescence lifetime imaging microscopy, and be applied on both in vivo and in vitro specimens. This opens significant opportunities in molecular imaging of metabolic activities, intracellular sensing, and trafficking as well as in vivo exploration of cell-tissue interfaces.  相似文献   

16.
Glycosylphosphatidylinositol-anchor biosynthesis and glycosylphosphatidylinositol modification of proteins are central to coordinated plant development.Since their discovery (Low and Saltiel, 1988), glycosylphosphatidylinositol-anchored proteins (GPI-APs) have provoked intense interest as crucial regulators for growth, morphogenesis, reproduction, and disease pathogenesis in organisms ranging from yeast and trypanosomes to animals and plants. The lipid moiety, the glycosylphosphatidylinositol (GPI) anchor, is synthesized in the endoplasmic reticulum (ER); the protein component is cotranslationally inserted into the ER and posttranslationally modified by the addition of a GPI anchor (Kinoshita et al., 2013; Fig. 1). GPI-APs are then transported via the Golgi to the outer surface of the plasma membrane. The lipid anchor mediates stable attachment of these proteins to the cell surface, where some play important roles as signaling regulators from sphingolipid- and sterol-enriched membrane microdomains (Simons and Gerl, 2010). Some GPI-APs are released from the cell membrane by phosphatidylinositol-specific phospholipases to the extracellular matrix, where they might engage in processes such as cell adhesion and cell-cell communication. In Arabidopsis (Arabidopsis thaliana), there are about 250 predicted GPI-APs (Borner et al., 2003), a relatively large number compared with about 150 in mammals and 50 in the budding yeast (Saccharomyces cerevisiae). Important functions for plant GPI-APs have been elucidated through the study of individual proteins, such as the COBRA family in cell expansion and cell wall biosynthesis (Brady et al., 2007), ARABINOGALACTAN PROTEIN18 in megagametogenesis (Demesa-Arévalo and Vielle-Calzada, 2013), and LORELEI in the pollen tube-female gametophyte interaction (Capron et al., 2008; Tsukamoto et al., 2010; Duan et al., 2014). However, it is the studies of mutants defective in GPI biosynthesis that underscore the general importance of GPI-APs as a class: lacking the capacity to assemble the anchor is lethal.Open in a separate windowFigure 1.GPI anchor biosynthesis pathway. Ten or 11 stepwise modifications of phosphoinositide occur, starting from the synthesis of N-glucosamine-phosphoinositide on the cytoplasmic surface of the ER, followed by its flipping to the ER lumenal side for additional modifications, ending with the addition of the terminal ethanolamine phosphate. Proteins destined for GPI modification are synthesized with a C-terminal signature sequence recognized by the GPI transamidase (a five-protein-enzyme complex) that concomitantly cleaves the peptide at what is designated as the ω and ω+1 amino acids and attaches the GPI anchor in a transamination reaction (red arrows). The GPI-modified proteins are then sorted and transported via the Golgi apparatus to the cell membrane. The established biosynthetic proteins from Arabidopsis and their mammalian homologs are indicated; the galactosylation step appears to be plant specific. The diagram is modeled after figure 3 in Ellis et al. (2010), which also provided a complete list of potential plant orthologs of the human and yeast proteins in the pathway.The GPI anchor is synthesized by an elaborate biosynthetic pathway, starting on the cytoplasmic side of the ER and ending with a completely assembled core anchor on the lumenal surface of the ER (Fig. 1). Prior to their transport out of the ER, proteins destined for GPI modification are cleaved at a C-terminal signature sequence by a GPI transamidase complex that in two enzymatic steps concomitantly attaches a GPI anchor to the C terminus of processed proteins (Kinoshita, 2014). Most of the knowledge on GPI biosynthesis and GPI-AP modification is derived from studies in mammals and yeast, but the pathway is likely to be conserved in plants (Ellis et al., 2010). In a recent article in Plant Physiology, Dai et al. (2014) reported that a GPI anchor biosynthesis mutant, abnormal pollen tube guidance1 (atpg1), displays both embryo lethality and severely depressed male fertility. They determined that APTG1 is homologous to the yeast GPI10 and human PIG-B (for phosphatidylinositol glycan anchor biosynthesis) proteins, the last of three distinct mannosyltransferases that modify the precursor anchor (Fig. 1), and showed that APTG1 can functionally substitute for GPI10 in a conditionally lethal gpi10 yeast mutant. Previous studies have demonstrated that Arabidopsis SETH1 (a male fertility god in Egyptian mythology), SETH2, and PEANUT1 (PNT1), encoding homologs of mammalian PIG-C, PIG-A, and PIG-M (Fig. 1) and their corresponding yeast counterparts, respectively, are important for male fertility (Lalanne et al., 2004; Gillmor et al., 2005). In addition, loss of the first mannosyltransferase in the pathway in pnt1 results in early seedling lethality. pnt1 embryos are severely defective, displaying various cell division anomalies and exhibiting altered levels and ectopic deposition of cell wall polymers. The results reported by Dai et al. (2014), therefore, further demonstrate the conservation of the GPI biosynthesis pathway and the importance of GPI anchoring in plant development and reproduction.The aptg1 mutant was isolated in a search for mutants defective in pollen tube targeting of ovules (Fig. 2), an intriguing and crucial step in plant reproduction. A pollen tube is guided to an ovule by attractants, and upon reaching the target, the female gametophyte, the pollen tube ruptures, ejecting its cytoplasm and releasing sperm for fertilization (Dresselhaus and Franklin-Tong, 2013). aptg1 pollen tubes either fail to target ovules or undertake a more twisted pathway before entering an ovule. In an earlier study, Li et al. (2013) showed that a GPI-AP, COBRA-LIKE10 (COBL10), is required to maintain normal pollen tube growth rates and ovule targeting efficiency. In aptg1 pollen tubes, citrine fluorescent protein-COBL10 was absent from its normal apical membrane location while the citrine fluorescent signal in the cytoplasm was more intense, implying that the diminished presence of COBL10 on the apical membrane could be an underlying cause for the ovule-targeting phenotype. This observation also demonstrates that GPI anchoring is important for the subsequent sorting and transport of these proteins to their destined locations (Kinoshita et al., 2013) and consistent with a wholesale failure of GPI-APs to reach their functional locations as underlying lethality in GPI biosynthesis mutants.Open in a separate windowFigure 2.Pollen tube targeting of ovules in an Arabidopsis pistil. GUS-expressing pollen grains pollinated the pistil. Each blue dot represents discharged cytoplasm from a pollen tube that, in response to attractants, has successfully targeted the ovule and penetrated the female gametophyte and was induced to burst. The cytoplasmic discharge releases sperm for fertilization.While it is clear that major biological roles are played by GPI-APs, many questions remain. Most constituents of the plant GPI anchor biosynthetic pathway remain to be functionally established (Fig. 1). Much has been said about the membrane environments where GPI-APs are localized, but we are far from understanding the precise roles they play in assembling these domains and regulating their functional dynamics. Advances in high-resolution imaging at the cell surface and biochemical approaches to determine the constituents in these membrane microdomains (Simons and Gerl, 2010) should accelerate our understanding of the importance of GPI anchoring as a conserved strategy among eukaryotes to control a wide range of processes.  相似文献   

17.
Riboflavin (vitamin B2) is the precursor of the flavin coenzymes flavin mononucleotide and flavin adenine dinucleotide. In Escherichia coli and other bacteria, sequential deamination and reduction steps in riboflavin biosynthesis are catalyzed by RibD, a bifunctional protein with distinct pyrimidine deaminase and reductase domains. Plants have two diverged RibD homologs, PyrD and PyrR; PyrR proteins have an extra carboxyl-terminal domain (COG3236) of unknown function. Arabidopsis (Arabidopsis thaliana) PyrD (encoded by At4g20960) is known to be a monofunctional pyrimidine deaminase, but no pyrimidine reductase has been identified. Bioinformatic analyses indicated that plant PyrR proteins have a catalytically competent reductase domain but lack essential zinc-binding residues in the deaminase domain, and that the Arabidopsis PyrR gene (At3g47390) is coexpressed with riboflavin synthesis genes. These observations imply that PyrR is a pyrimidine reductase without deaminase activity. Consistent with this inference, Arabidopsis or maize (Zea mays) PyrR (At3g47390 or GRMZM2G090068) restored riboflavin prototrophy to an E. coli ribD deletant strain when coexpressed with the corresponding PyrD protein (At4g20960 or GRMZM2G320099) but not when expressed alone; the COG3236 domain was unnecessary for complementing activity. Furthermore, recombinant maize PyrR mediated NAD(P)H-dependent pyrimidine reduction in vitro. Import assays with pea (Pisum sativum) chloroplasts showed that PyrR and PyrD are taken up and proteolytically processed. Ablation of the maize PyrR gene caused early seed lethality. These data argue that PyrR is the missing plant pyrimidine reductase, that it is plastid localized, and that it is essential. The role of the COG3236 domain remains mysterious; no evidence was obtained for the possibility that it catalyzes the dephosphorylation that follows pyrimidine reduction.Riboflavin is the substrate for biosynthesis of the essential flavocoenzymes FMN and FAD, which occur in all kingdoms of life and have roles in diverse redox reactions as well as in other processes such as DNA repair, light sensing, and bioluminescence (Fischer and Bacher, 2005). Plants and many microorganisms can synthesize riboflavin, but humans and other animals cannot, so they must obtain it from the diet (Powers, 2003). Plant foods are important sources of riboflavin for humans, and the riboflavin pathway is a target for engineering biofortified crops (Fitzpatrick et al., 2012).Riboflavin biosynthesis proceeds via the same pathway in bacteria and plants (Fischer and Bacher, 2005; Roje, 2007). This pathway starts from GTP, which is converted by GTP cyclohydrolase II (named RibA in Escherichia coli) to the pyrimidine derivative 2,5-diamino-6-ribosylamino-4(3H)-pyrimidinone 5′-P. Deamination of the pyrimidine ring then yields 5-amino-6-ribosylamino-2,4(1H,3H)-pyrimidinedione 5′-P, and subsequent reduction of the ribosyl moiety gives 5-amino-6-ribitylamino-2,4(1H,3H)-pyrimidinedione 5′-P. After dephosphorylation, this product is condensed with 3,4-dihydroxy-2-butanone 4-P to give 6,7-dimethyl-8-ribityllumazine, whose dismutation yields riboflavin. Figure 1 shows the first four steps of this pathway.Open in a separate windowFigure 1.The first four steps of the riboflavin biosynthesis pathway in bacteria and plants. The enzymes involved are GTP cyclohydrolase II (RibA), pyrimidine deaminase (Deam), pyrimidine reductase (Red), and a specific phosphatase (Pase). Enzymes for which the plant genes are not known are colored red. Intermediates are as follows: 1, 2,5-diamino-6-ribosylamino-4(3H)-pyrimidinone 5′-P; 2, 5-amino-6-ribosylamino-2,4(1H,3H)-pyrimidinedione 5′-P; 3, 5-amino-6-ribitylamino-2,4(1H,3H)-pyrimidinedione 5′-P; 4, 5-amino-6-ribitylamino-2,4(1H,3H)-pyrimidinedione.In E. coli, the deamination and reduction steps are catalyzed by a single bifunctional enzyme, RibD, which has N-terminal deaminase and C-terminal reductase domains that retain their respective activities when expressed separately (Richter et al., 1997; Magalhães et al., 2008). The situation in plants seems superficially similar but is in fact more complex (Gerdes et al., 2012). The bidomain bacterial RibD protein has two types of homologs in plants (Fischer et al., 2004; Chatwell et al., 2006; Chen et al., 2006), here called PyrD and PyrR, both with apparent deaminase and reductase domains (Fig. 2A). Only PyrD, represented by At4g20960, has been studied biochemically; it was found to have pyrimidine deaminase but not reductase activity (Fischer et al., 2004). The function of PyrR, represented by At3g47390, is unknown, although it has been inferred to have reductase activity (Chatwell et al., 2006; Chen et al., 2006; Ouyang et al., 2010) and perhaps to lack deaminase activity (Ouyang et al., 2010). Another mystery surrounding PyrR proteins is the presence of an extra C-terminal domain of unknown function (COG3236 in the Clusters of Orthologous Groups database; Fig. 2A); this domain occurs as a stand-alone protein in many bacteria. One possibility is that it catalyzes the dephosphorylation that follows the pyrimidine reduction step in the pathway (Fig. 1). The phosphatase involved is most likely substrate specific, but it has not been identified in plants or any other organism (Roje, 2007; Gerdes et al., 2012), and genes for enzymes in the same pathway, especially for successive steps, are quite commonly fused (Suhre, 2007). A mutation (phs1) that deleted the COG3236 domain from Arabidopsis (Arabidopsis thaliana) PyrR resulted in a photosensitive phenotype that could be rescued by supplied FAD (Ouyang et al., 2010).Open in a separate windowFigure 2.Structure and phylogeny of plant PyrD and PyrR proteins. A, Domain architectures. The examples shown are Arabidopsis At4g20960 and At3g47390; the predicted plastid targeting peptide (TP) varies in length between species. B, Phylogenetic tree of PyrD and PyrR proteins. Sequences were aligned with ClustalW; the tree was built by the neighbor-joining method with MEGA5. Bootstrap values (%) for 1,000 replicates are next to the nodes. Only the tree topology is shown. Note that the PyrD proteins of green algae (underlined) lack a reductase domain. C, Alignments showing the conservation of the zinc-binding residues (arrowheads) in the deaminase domain of PyrD but not PyrR proteins and the conservation of the predicted substrate-binding residues (asterisks) in the reductase domain of PyrR but not PyrD proteins. The deaminase sequences correspond to residues 45 to 85 of B. subtilis RibD (synonym RibG); the reductase sequences correspond to residues 150 to 210 and (separated by dots) 288 to 292 of B. subtilis RibD. Identical zinc- or substrate-binding residues are black, and conservative replacements are gray. Dashes indicate gaps that maximize the alignment.The plant riboflavin synthesis pathway is considered to be plastidial (Roje, 2007), but this location is based almost solely on bioinformatics and high-throughput proteome analyses (Gerdes et al., 2012). In only one case is there more definitive experimental support: in vitro chloroplast import data for the pathway’s penultimate enzyme, 6,7-dimethyl-8-ribityllumazine synthase (Jordan et al., 1999). Similarly, clear genetic support for the function of most plant riboflavin synthesis enzymes is lacking (Gerdes et al., 2012), the exception being an Arabidopsis RibA homolog (Hedtke and Grimm, 2009).The work reported here established, using maize (Zea mays) and Arabidopsis, that PyrR is indeed the missing pyrimidine reductase, that it lacks deaminase activity, and that its COG3236 domain is not essential for pyrimidine reductase activity and most likely lacks phosphatase activity. We also demonstrated the import of PyrR and PyrD into chloroplasts in vitro and confirmed that the gene for PyrR is essential.  相似文献   

18.
Two nonoverlapping autosomal inversions defined unusual neo-sex chromosomes in the Hessian fly (Mayetiola destructor). Like other neo-sex chromosomes, these were normally heterozygous, present only in one sex, and suppressed recombination around a sex-determining master switch. Their unusual properties originated from the anomalous Hessian fly sex determination system in which postzygotic chromosome elimination is used to establish the sex-determining karyotypes. This system permitted the evolution of a master switch (Chromosome maintenance, Cm) that acts maternally. All of the offspring of females that carry Cm-associated neo-sex chromosomes attain a female-determining somatic karyotype and develop as females. Thus, the chromosomes act as maternal effect neo-W''s, or W-prime (W′) chromosomes, where ZW′ females mate with ZZ males to engender female-producing (ZW′) and male-producing (ZZ) females in equal numbers. Genetic mapping and physical mapping identified the inversions. Their distribution was determined in nine populations. Experimental matings established the association of the inversions with Cm and measured their recombination suppression. The inversions are the functional equivalent of the sciarid X-prime chromosomes. We speculate that W′ chromosomes exist in a variety of species that produce unisexual broods.SEX chromosomes are usually classified as X, Y, Z, or W on the basis of their pattern of segregation and the gender of the heterogametic sex (Ohno 1967). However, when chromosome-based sex determination occurs postzygotically, the same nomenclature confounds important distinctions and may hide interesting evolutionary phenomena. The Hessian fly (Mayetiola destructor), a gall midge (Diptera: Cecidomyiidae) and an important insect pest of wheat, presents an excellent example (Stuart and Hatchett 1988, 1991). In this insect, all of the female gametes and all of the male gametes have the same number of X chromosomes (Figure 1A); no heterogametic sex exists. Nevertheless, Hessian fly sex determination is chromosome based; postzygotic chromosome elimination produces different X chromosome to autosome ratios in somatic cells (male A1A2X1X2/A1A2OO and female A1A2X1X2/A1A2X1X2, where A1 and A2 are the autosomes, X1 and X2 are the X chromosomes, and the paternally derived chromosomes follow the slash) (Stuart and Hatchett 1991; Marin and Baker 1998). Thus, Hessian fly “X” chromosomes are defined by their haploid condition in males, rather than by their segregation in the gametes.Open in a separate windowFigure 1.—Chromosome behavior and sex determination in the Hessian fly. (A) Syngamy (1) establishes the germ-line chromosome constitution: ∼32 maternally derived E chromosomes (represented as a single white chromosome) and both maternally derived (black) and paternally derived (gray) autosomes and X chromosomes. During embryogenesis, while the E chromosomes are eliminated, the paternally derived X chromosomes are either retained (2) or excluded (3) from the presumptive somatic cells. When the paternally derived X chromosomes are retained (2), a female-determining karyotype is established. When they are eliminated (3), a male-determining karyotype is established. Thelygenic mothers carry Cm (white arrow), which conditions all of their offspring to retain the X chromosomes. Recombination occurs during oogenesis (4). All ova contain a full complement of E chromosomes and a haploid complement of autosomes and X chromosomes. Chromosome elimination occurs during spermatogenesis (5). Sperm contain only the maternally derived autosomes and X chromosomes. (B) The segregation of Cm (white dot) on a Hessian fly autosome among monogenic families. Thelygenic females produce broods composed of equal numbers of thelygenic (Cm/−) and arrhenogenic (−/−) females (box 1). Arrhenogenic females produce males (box 2). (C) Matings between monogenic and amphigenic families. Cm (white dot) is dominant to the amphigenic-derived chromosomes (gray dot) and generates all-female offspring (box 3). Amphigenic-derived chromosomes are dominant to the arrhenogenic-derived chromosomes (no dot) and generate offspring of both sexes (box 4).An autosomal, dominant, genetic factor called Chromosome maintenance (Cm) complicates Hessian fly sex determination further (Stuart and Hatchett 1991). Cm has a maternal effect that acts upstream of X chromosome elimination during embryogenesis (Figure 1A). It prevents X chromosome elimination so that all of the offspring of Cm-bearing mothers obtain a female-determining karyotype. Cm-bearing females produce only female offspring and are therefore thelygenic. The absence of Cm usually has the opposite effect; all of the offspring of most Cm-lacking females obtain a male-determining karyotype. These Cm-lacking females produce only male offspring and are therefore arrhenogenic. Like a sex-determining master switch, Cm is usually heterozygous and present in only one sex (Figure 1B). Thus, thelygenic females (Cm/−) are “heterogametic,” as their Cm-containing gametes and Cm-lacking gametes produce thelygenic (Cm/−) and arrhenogenic (−/−) females in a 1:1 ratio. Collectively, thelygenic and arrhenogenic females are called monogenic because they produce unisexual families. However, some Hessian fly females produce broods of both sexes and are called amphigenic. No mating barrier between monogenic and amphigenic families exists (Figure 1C), but amphigenic females have always been found in lower abundance (Painter 1930; Gallun et al. 1961; Stuart and Hatchett 1991). In experimental matings, the inheritance of maternal phenotype was consistent with the segregation of three Cm alleles (Figure 1C): a dominant thelygenic allele, a hypomorphic amphigenic allele, and a null arrhenogenic allele (Stuart and Hatchett 1991).Here we report the genetic and physical mapping of Cm on Hessian fly autosome 1 (A1). Two nonoverlapping inversions were identified that segregated perfectly with Cm. The most distal inversion was present in all thelygenic females examined. The more proximal inversion extended recombination suppression. These observations suggested that successive inversions evolved to suppress recombination around Cm after it arose. The inversions therefore appear to have evolved in response to the forces that shaped vertebrate Y and W chromosomes (Charlesworth 1996; Graves and Shetty 2001; Rice and Chippindale 2001; Carvalho and Clark 2005). We therefore believe the inversion-bearing chromosomes may be classified as maternal effect neo-W''s.  相似文献   

19.
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   

20.
Radioisotope-based and mass spectrometry coupled to chromatographic techniques are the conventional methods for monitoring HMG-CoA reductase (HMGR) activity. Irrespective of offering adequate sensitivity, these methods are often cumbersome and time-consuming, requiring the handling of radiolabeled chemicals or elaborate ad-hoc derivatizing procedures. We propose a rapid and versatile reverse phase-HPLC method for assaying HMGR activity capable of monitoring the levels of both substrates (HMG-CoA and NADPH) and products (CoA, mevalonate, and NADP+) in a single 20 min run with no pretreatment required. The linear dynamic range was 10–26 pmol for HMG-CoA, 7–27 nmol for NADPH, 0.5–40 pmol for CoA and mevalonate, and 2–27 nmol for NADP+, and limit of detection values were 2.67 pmol, 2.77 nmol, 0.27 pmol, and 1.3 nmol, respectively.HMG-CoA reductase (HMGR) is the enzyme that catalyze the four-electron reductive deacylation of HMG-CoA to CoA and mevalonate (Fig. 1) (1). This reaction is the controlling step in the biosynthesis of sterols and isoprenoids (2, 3); hence, a large number of studies on the modulation of HMGR activity are continuously performed in the effort of developing new drugs in the treatment of hypercholesterolemic disorders (1).Open in a separate windowFig. 1.Schematic representation of HMGR enzymatic reaction.HMGR activity is conventionally assayed using elaborate radiochemical assay (49), chromatographic techniques coupled with mass spectrometry (1015), or spectrophotometrically by monitoring the decrease in the absorbance of cofactor NADPH at 340 nm (16).Herein, as an alternative for laboratories with no access to the expensive LC/MS equipment, we propose a rapid and adequately sensitive HPLC-based method capable of monitoring both the levels of all the species involved in the equilibrium in a single analysis and the kinetics of HMGR-catalyzed reactions.  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