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.
2.
3.
This study demonstrates the utility of Lifeact for the investigation of actin dynamics in Neurospora crassa and also represents the first report of simultaneous live-cell imaging of the actin and microtubule cytoskeletons in filamentous fungi. Lifeact is a 17-amino-acid peptide derived from the nonessential Saccharomyces cerevisiae actin-binding protein Abp140p. Fused to green fluorescent protein (GFP) or red fluorescent protein (TagRFP), Lifeact allowed live-cell imaging of actin patches, cables, and rings in N. crassa without interfering with cellular functions. Actin cables and patches localized to sites of active growth during the establishment and maintenance of cell polarity in germ tubes and conidial anastomosis tubes (CATs). Recurrent phases of formation and retrograde movement of complex arrays of actin cables were observed at growing tips of germ tubes and CATs. Two populations of actin patches exhibiting slow and fast movement were distinguished, and rapid (1.2 μm/s) saltatory transport of patches along cables was observed. Actin cables accumulated and subsequently condensed into actin rings associated with septum formation. F-actin organization was markedly different in the tip regions of mature hyphae and in germ tubes. Only mature hyphae displayed a subapical collar of actin patches and a concentration of F-actin within the core of the Spitzenkörper. Coexpression of Lifeact-TagRFP and β-tubulin–GFP revealed distinct but interrelated localization patterns of F-actin and microtubules during the initiation and maintenance of tip growth.Actins are highly conserved proteins found in all eukaryotes and have an enormous variety of cellular roles. The monomeric form (globular actin, or G-actin) can self-assemble, with the aid of numerous actin-binding proteins (ABPs), into microfilaments (filamentous actin, or F-actin), which, together with microtubules, form the two major components of the fungal cytoskeleton. Numerous pharmacological and genetic studies of fungi have demonstrated crucial roles for F-actin in cell polarity, exocytosis, endocytosis, cytokinesis, and organelle movement (6, 7, 20, 34, 35, 51, 52, 59). Phalloidin staining, immunofluorescent labeling, and fluorescent-protein (FP)-based live-cell imaging have revealed three distinct subpopulations of F-actin-containing structures in fungi: patches, cables, and rings (1, 14, 28, 34, 60, 63, 64). Actin patches are associated with the plasma membrane and represent an accumulation of F-actin around endocytic vesicles (3, 26, 57). Actin cables are bundles of actin filaments stabilized with cross-linking proteins, such as tropomyosins and fimbrin, and are assembled by formins at sites of active growth, where they form tracks for myosin V-dependent polarized secretion and organelle transport (10, 16, 17, 27, 38, 47, 48). Cables, unlike patches, are absolutely required for polarized growth in the budding yeast Saccharomyces cerevisiae (34, 38). Contractile actomyosin rings are essential for cytokinesis in budding yeast, whereas in filamentous fungi, actin rings are less well studied but are known to be involved in septum formation (20, 28, 34, 39, 40).Actin cables and patches have been particularly well studied in budding yeast. However, there are likely to be important differences between F-actin architecture and dynamics in budding yeast and those in filamentous fungi, as budding yeasts display only a short period of polarized growth during bud formation, which is followed by isotropic growth over the bud surface (10). Sustained polarized growth during hyphal morphogenesis is a defining feature of filamentous fungi (21), making them attractive models for studying the roles of the actin cytoskeleton in cell polarization, tip growth, and organelle transport.In Neurospora crassa and other filamentous fungi, disruption of the actin cytoskeleton leads to rapid tip swelling, which indicates perturbation of polarized tip growth, demonstrating a critical role for F-actin in targeted secretion to particular sites on the plasma membrane (7, 22, 29, 56). Immunofluorescence studies of N. crassa have shown that F-actin localizes to hyphal tips as “clouds” and “plaques” (7, 54, 59). However, immunolabeling has failed to reveal actin cables in N. crassa and offers limited insights into F-actin dynamics. Live-cell imaging of F-actin architecture and dynamics has not been accomplished in N. crassa, yet it is expected to yield key insights into cell polarization, tip growth, and intracellular transport.We took advantage of a recently developed live-cell imaging probe for F-actin called Lifeact (43). Lifeact is a 17-amino-acid peptide derived from the N terminus of the budding yeast actin-binding protein Abp140 (5, 63) and has recently been demonstrated to be a universal live-cell imaging marker for F-actin in eukaryotes (43). Here, we report the successful application of fluorescent Lifeact fusion constructs for live-cell imaging of F-actin in N. crassa. We constructed two synthetic genes consisting of Lifeact fused to “synthetic” green fluorescent protein (sGFP) (S65T) (henceforth termed GFP) (12) or red fluorescent protein (TagRFP) (33) and expressed these constructs in various N. crassa strains. In all strain backgrounds, fluorescent Lifeact constructs clearly labeled actin patches, cables, and rings and revealed a direct association of F-actin structures with sites of cell polarization and active tip growth. Our results demonstrate the efficacy of Lifeact as a nontoxic live-cell imaging probe in N. crassa.  相似文献   

4.
5.
6.
We recently reported that induced pluripotent stem cells (iPSCs) prepared from different human origins acquired similar glycan profiles to one another as well as to human embryonic stem cells. Although the results strongly suggested attainment of specific glycan expressions associated with the acquisition of pluripotency, the detailed glycan structures remained to be elucidated. Here, we perform a quantitative glycome analysis targeting both N- and O-linked glycans derived from 201B7 human iPSCs and human dermal fibroblasts as undifferentiated and differentiated cells, respectively. Overall, the fractions of high mannose-type N-linked glycans were significantly increased upon induction of pluripotency. Moreover, it became evident that the type of linkage of Sia on N-linked glycans was dramatically changed from α-2–3 to α-2–6, and the expression of α-1–2 fucose and type 1 LacNAc structures became clearly apparent, while no such glycan epitopes were detected in fibroblasts. The expression profiles of relevant glycosyltransferase genes were fully consistent with these results. These observations indicate unambiguously the manifestation of a “glycome shift” upon conversion to iPSCs, which may not merely be the result of the initialization of gene expression, but could be involved in a more aggressive manner either in the acquisition or maintenance of the undifferentiated state of iPSCs.Induced pluripotent stem cells (iPSCs)1 are genetically manufactured pluripotent cells obtained by the transfection of reprogramming factors. Such iPSCs were first reported in 2006 for the mouse (1) and in 2007 for humans (2, 3). Although iPSCs have already been used in the fields of drug development and disease models (47), basic aspects of iPSCs largely remain to be elucidated to provide us with a fuller understanding of their properties and for therapeutic applications to be developed in the field of regenerative medicine. These aspects include the need for a definitive system to be established to evaluate their properties; e.g. pluripotency, differentiation propensity, risk of possible contamination of xenoantigens, and even the potential for tumorigenesis. Cell surface glycans are often referred to as the “cell signature,” which changes dramatically depending on the cell properties and conditions (8) as a result of changes in gene expression, including epigenetic modifications of glycan-related molecules. Glycans, because of their outermost cell-surface locations and structural complexity, are considered to be most advantageous communication molecules, playing roles in various biological phenomena. Indeed, SSEA3/4 and Tra-1–60/81, which have been used to discriminate pluripotency, are cell surface glycan epitopes that respond to some specific antibodies (912).Glycan-mediated cell-to-cell interactions have been shown to play important roles in various biological phenomena including embryogenesis and carcinogenesis (1316). This might also be the case for the acquisition and maintenance of iPSC and ESC pluripotency, although there remains much to clarify concerning the roles of cell surface glycans in these events. Thus, the development of novel cell surface markers to evaluate the properties of iPSCs and ESCs is keenly required. Toward this goal, a glycomic approach has been made by several groups (1720). In our previous study using an advanced lectin microarray technique (21), thirty-eight lectins capable of discriminating between iPSCs and SCs were statistically selected, and the characteristic features of the pluripotent state were obtained. The glycan profiles of the parent SCs, derived from four different tissues, were totally different from one another and from those of the iPSCs. Despite this observation, the technique used lacks the ability to determine detailed glycan structures or allow their quantification. For this purpose, a conventional approach based on high performance liquid chromatography (HPLC) combined with matrix-assisted laser desorption-ionization (MALDI) - time of flight (TOF) mass spectrometry (MS) was undertaken for both the definitive identification of glycan structures and their quantitative comparison, which remained unclear in the previous analysis (21).We report here structural data on N-linked and O-linked glycans derived from the human iPSC 201B7 cell line (2) and human dermal fibroblasts (SC) representing undifferentiated and differentiated cells, respectively. For quantitative comparison, the glycans were liberated by gas-phase hydrazinolysis from similar numbers of cells (2225) fluorescently tagged with 2-aminopyridine (2-AP) at their reducing terminus (26, 27), following which the derived pyridylaminated (PA-) glycans were purified by multiple-mode (i.e. anion-exchange, size-fractionation and reverse-phase) HPLC. Their structures were determined and quantified by HPLC mapping assisted with MALDI-TOF-MS and exoglycosidase digestion analyses. This report thus provides the first structural evidence showing the occurrence of a dynamic “glycome shift” upon induction of pluripotency.  相似文献   

7.
Tetrahydrolipstatin (THL) is bactericidal but its precise target spectrum is poorly characterized. Here, we used a THL analog and activity-based protein profiling to identify target proteins after enrichment from whole cell lysates of Mycobacterium bovis Bacillus Calmette-Guérin cultured under replicating and non-replicating conditions. THL targets α/β-hydrolases, including many lipid esterases (LipD, G, H, I, M, N, O, V, W, and TesA). Target protein concentrations and total esterase activity correlated inversely with cellular triacylglycerol upon entry into and exit from non-replicating conditions. Cellular overexpression of lipH and tesA led to decreased THL susceptibility thus providing functional validation. Our results define the target spectrum of THL in a biological species with particularly diverse lipid metabolic pathways. We furthermore derive a conceptual approach that demonstrates the use of such THL probes for the characterization of substrate recognition by lipases and related enzymes.Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), is responsible for nearly 2 million deaths each year. The host immune response toward aerosol infection is to quarantine tubercle bacilli in a granulomatous structure (1, 2). However, granuloma-associated mycobacteria can switch to a non-replicative, “dormant” state and successfully evade immune response for decades after infection (3, 4). The metabolic events that permit tubercle bacilli to enter host cells and revive from states of persistence suggest that lipids are utilized as a carbon source (57). During times of oxygen deprivation and in the absence of host cells, cultivated mycobacteria store fatty acids (FAs) in the form of triacylglycerol (TAG)1-enriched lipid droplets (810). Upon resuscitation (by the re-introduction of oxygen), these lipid droplets vanish and TAGs are hydrolyzed (11). Unfortunately, the molecular mechanisms for TAG build-up and breakdown are far less well understood in bacteria when compared with those processes in eukaryotes.Comparative sequence analysis of the Mtb genome has revealed that it contains 250 genes encoding enzymes involved in lipid metabolism compared with only 50 enzymes in Escherichia coli, which has a genome of comparable size. Among these genes, 150 are predicted to encode proteins involved in lipid catabolism (12, 13). A family of 24 carboxyl ester hydrolases called “lip” genes (lipC to Z, except K and S) has been predicted to play a role in lipid catabolism (14). Among these, only a few have been functionally characterized and related to mycobacterial dormancy and resuscitation (1518).Tetrahydrolipstatin, a serine esterase inhibitor, covalently binds to and inhibits mammalian lipases and fatty acid synthase (FAS) and is marketed as “Orlistat” for the treatment of severe forms of obesity (19). THL was previously shown to inhibit both active and latent forms of mycobacteria (11, 2022) but the bacterial target spectrum remains poorly characterized. Therefore, to (1) define the THL target spectrum in a mycobacterial species and (2) to obtain biochemical insights into regulation of lipases and esterases in different metabolic states, we employed a chemical-proteomics approach using activity-based protein profiling (ABPP) with a bait that has been described to bind to lipolytic enzymes (2325). We identified several known lipases (as anticipated), putative lipase and esterases, and hypothetical proteins of unknown functions, thereby providing a comprehensive resource of experimentally determined THL targets in mycobacteria. Importantly, we systematically compared readouts of fluorescently tagged THL-proteins (7 bands on one-dimensional SDS-PAGE) with those of mass spectrometry-based peptide identification of enriched protein fractions (247 in growing cells). This comparison led to the identification of 14 THL targets, two of which were further validated experimentally. We furthermore provide a conceptual framework for the evaluation of this target list using both experimental as well as bioinformatics approaches in two examples, lipH and tesA. Overall, our data indicate that THL is an anti-mycobacterial drug because of its potential to (1) bind to a relatively wide range of lipolytic enzymes and (2) prevent bacilli from resuscitating from a nonreplicating persistent (NRP) state when lipid metabolism is particularly important.  相似文献   

8.
9.
The N-glycosylation of the model nematode Caenorhabditis elegans has proven to be highly variable and rather complex; it is an example to contradict the existing impression that “simple” organisms possess also a rather simple glycomic capacity. In previous studies in a number of laboratories, N-glycans with up to four fucose residues have been detected. However, although the linkage of three fucose residues to the N,N′-diacetylchitobiosyl core has been proven by structural and enzymatic analyses, the nature of the fourth fucose has remained uncertain. By constructing a triple mutant with deletions in the three genes responsible for core fucosylation (fut-1, fut-6 and fut-8), we have produced a nematode strain lacking products of these enzymes, but still retaining maximally one fucose residue on its N-glycans. Using mass spectrometry and HPLC in conjunction with chemical and enzymatic treatments as well as NMR, we examined a set of α-mannosidase-resistant N-glycans. Within this glycomic subpool, we can reveal that the core β-mannose can be trisubstituted and so carries not only the ubiquitous α1,3- and α1,6-mannose residues, but also a “bisecting” β-galactose, which is substoichiometrically modified with fucose or methylfucose. In addition, the α1,3-mannose can also be α-galactosylated. Our data, showing the presence of novel N-glycan modifications, will enable more targeted studies to understand the biological functions and interactions of nematode glycans.Nematodes represent, along with arthropods, one of the largest groups of animals to exist on the planet; 25.000 species are described, but the existence of up to one million has been estimated (1, 2). They have various ecological niches and include free-living “worms” in the soil, fungivorous, entomopathogenic, and necromenic species as well as parasites of plants and mammals, which share the basic conserved body plan (more-or-less a digestive tube surrounded with muscle, whether larger or smaller). There are five major clades (Rhabditina, Enoplia, Spirurina, Tylenchina, and Dorylaimia) (2), yet the glycosylation of only a few nematode species has been studied with an inevitable focus on the model nematode Caenorhabditis elegans and parasitic species (3). Thereby, the use of C. elegans mutants has been highly valuable in dissecting aspects of nematode N-glycan biosynthesis and revealing the in vivo substrates for certain glycosyltransferases (4).As many nematodes are parasites, their interactions with the immune systems of their hosts have attracted attention; particularly, there are relationships between autoimmunity, allergy, vaccination, and helminth infections. The “old friends” hypothesis seeks to understand the evolutionary factors that have shaped the immune system and to explain correlations between lifestyles in the developed world and modern “epidemics,” which are due to immunological misbalance (57). Promising data have suggested that “worm therapy” may bring advantages to some patients with Crohn''s disease or allergies (8, 9); however, such approaches are controversial. Nevertheless, crude extracts even of Caenorhabditis elegans were shown to induce a glycan-dependent Th2 response (10), whereas the excretory-secretory products of some nematodes also have immunomodulatory activity (11). Furthermore, the native glycoproteins of some nematodes have proven effective in vaccination trials, whereas recombinant forms are not, which is suggestive that post-translational modifications may have a role in an efficacious immune response (12).As at least some of the molecules relevant to nematode immunomodulation or vaccination are glycoproteins, a proper understanding of nematode glycosylation is of biomedical and veterinary relevance. Over the years, it has become apparent that the core chitobiosyl region of nematode N-glycans is subject to a range of modifications, with up to three core fucose residues being present (α1,3- and α1,6-linked on the reducing-terminal “proximal” GlcNAc and α1,3-linked on the second “distal” GlcNAc). However, up to four fucose residues have been detected on C. elegans N-glycans and the exact nature of the linkage of the fourth fucose has remained obscure despite work in our own and other laboratories (3, 1315). Combined with the latest knowledge regarding the specificity of C. elegans core fucosyltransferases (13, 16, 17) as well as our recent data regarding the exact structures of N-glycans from the C. elegans double hexosaminidase mutant and other nematodes (1820), we concluded that some models for the tri- and tetrafucosylated N-glycans were incorrect. By preparing a triple mutant unable to core fucosylate its N-glycans, we generated a C. elegans strain containing maximally one fucose residue on the N-linked oligosaccharides. Thereby a pool of unusual mannosidase-resistant N-glycans was identified and, using mass spectrometry (MS) and NMR, we reveal their modification with bisecting galactose frequently capped with fucose or methylfucose.  相似文献   

10.
Pyridine nucleotide transhydrogenase (PNT) catalyzes the direct transfer of a hydride-ion equivalent between NAD(H) and NADP(H) in bacteria and the mitochondria of eukaryotes. PNT was previously postulated to be localized to the highly divergent mitochondrion-related organelle, the mitosome, in the anaerobic/microaerophilic protozoan parasite Entamoeba histolytica based on the potential mitochondrion-targeting signal. However, our previous proteomic study of isolated phagosomes suggested that PNT is localized to organelles other than mitosomes. An immunofluorescence assay using anti-E. histolytica PNT (EhPNT) antibody raised against the NADH-binding domain showed a distribution to the membrane of numerous vesicles/vacuoles, including lysosomes and phagosomes. The domain(s) required for the trafficking of PNT to vesicles/vacuoles was examined by using amoeba transformants expressing a series of carboxyl-terminally truncated PNTs fused with green fluorescent protein or a hemagglutinin tag. All truncated PNTs failed to reach vesicles/vacuoles and were retained in the endoplasmic reticulum. These data indicate that the putative targeting signal is not sufficient for the trafficking of PNT to the vesicular/vacuolar compartments and that full-length PNT is necessary for correct transport. PNT displayed a smear of >120 kDa on SDS-PAGE gels. PNGase F and tunicamycin treatment, chemical degradation of carbohydrates, and heat treatment of PNT suggested that the apparent aberrant mobility of PNT is likely attributable to its hydrophobic nature. PNT that is compartmentalized to the acidic compartments is unprecedented in eukaryotes and may possess a unique physiological role in E. histolytica.Pyridine nucleotide transhydrogenase (PNT) participates in the bioenergetic processes of the cell. PNT generally resides on the cytoplasmic membranes of bacteria and the inner membrane of mammalian mitochondria (3, 16) and utilizes the electrochemical proton gradient across the membrane to drive NADPH formation from NADH (14, 15, 39) according to the reaction H+out + NADH + NADP+↔H+in + NAD+ + NADPH, where “out” and “in” denote the cytosol and the matrix of the mitochondria, or the periplasmic space and the cytosol of bacteria, respectively.PNT has been identified in several protozoan parasites, including Entamoeba histolytica (8, 51), Eimeria tenella (17, 47), Mastigamoeba balamuthi (11) Plasmodium falciparum (10), Plasmodium yoelii (6), and Plasmodium berghei (12). In general, PNT contains conserved structural units consisting of three domains, the NAD(H)-binding domain (domain I [dI]) and the NADP(H)-binding domain (domain III [dIII]), both of which face the matrix side of the eukaryotic mitochondria or the cytoplasmic side in bacteria, and the hydrophobic domain (domain II [dII]), containing 11 to 13 transmembrane regions. PNT from E. tenella and E. histolytica exists as a single polypeptide in an unusual configuration consisting of dIIb-dIII-dI-dIIa, with a 38-amino-acid-long linker region between dIII and dI (48).E. histolytica, previously considered an “amitochondriate” protist, is currently considered to possess a mitochondrion-related organelle with reduced and divergent functions, the mitosome (1, 21, 23a, 26, 42). Our recent proteomic study of isolated mitosomes identified about 20 new constituents (26), together with four proteins previously demonstrated in E. histolytica mitosomes: Cpn60 (8, 19, 21, 42), Cpn10 (46), mitochondrial Hsp70 (2, 44), and mitochondrion carrier family (MCF) (ADP/ATP transporter) (7). Despite the early presumption of PNT being localized in mitosomes (8), based on the amino-terminal region rich in hydroxylated (five serines and threonines) and acidic (three glutamates) amino acids, which slightly resembles known mitochondrion- and hydrogenosome-targeting sequences (8, 35), PNT was not discovered in the mitosome proteome. We also doubted this premise because PNT was one of the major proteins identified in isolated phagosomes (32, 33). Thus, the intracellular localization and trafficking of PNT remain unknown.In this report, we showed that E. histolytica PNT (EhPNT) is localized to various vesicles and vacuoles, including lysosomes and phagosomes, using wild-type amoebae and antiserum raised against recombinant EhPNT and an E. histolytica line expressing EhPNT with a carboxyl-terminal hemagglutinin (HA) epitope tag and anti-HA antibody. We also showed that all domains of EhPNT are required for its trafficking to the acidic compartment by using amoeba transformants expressing the HA tag or green fluorescent protein (GFP) fused with a region containing various domains of EhPNT.  相似文献   

11.
12.
The Aspergillus nidulans endocytic internalization protein SlaB is essential, in agreement with the key role in apical extension attributed to endocytosis. We constructed, by gene replacement, a nitrate-inducible, ammonium-repressible slaB1 allele for conditional SlaB expression. Video microscopy showed that repressed slaB1 cells are able to establish but unable to maintain a stable polarity axis, arresting growth with budding-yeast-like morphology shortly after initially normal germ tube emergence. Using green fluorescent protein (GFP)-tagged secretory v-SNARE SynA, which continuously recycles to the plasma membrane after being efficiently endocytosed, we establish that SlaB is crucial for endocytosis, although it is dispensable for the anterograde traffic of SynA and of the t-SNARE Pep12 to the plasma and vacuolar membrane, respectively. By confocal microscopy, repressed slaB1 germlings show deep plasma membrane invaginations. Ammonium-to-nitrate medium shift experiments demonstrated reversibility of the null polarity maintenance phenotype and correlation of normal apical extension with resumption of SynA endocytosis. In contrast, SlaB downregulation in hyphae that had progressed far beyond germ tube emergence led to marked polarity maintenance defects correlating with deficient SynA endocytosis. Thus, the strict correlation between abolishment of endocytosis and disability of polarity maintenance that we report here supports the view that hyphal growth requires coupling of secretion and endocytosis. However, downregulated slaB1 cells form F-actin clumps containing the actin-binding protein AbpA, and thus F-actin misregulation cannot be completely disregarded as a possible contributor to defective apical extension. Latrunculin B treatment of SlaB-downregulated tips reduced the formation of AbpA clumps without promoting growth and revealed the formation of cortical “comets” of AbpA.Germinating asexual spores (conidiospores) of Aspergillus nidulans transiently undergo isotropic growth (“swelling”) before establishing a polarity axis that grows by apical extension, leading to the characteristic tubular morphology of the fungal cell (15, 16, 33). Stable maintenance of a polarity axis at the high apical extension rates of A. nidulans (∼0.5 μm/min at 25°C) (23) can be attributable, at least in part, to the polarization of the secretory apparatus and the predominant and highly efficient delivery of secretory vesicles to the apex (8, 18, 40, 49). In addition, work from several laboratories strongly indicated that hyphal tip growth also involves endocytosis. A key observation supporting this involvement was that despite the fact that endocytosis can occur elsewhere, the endocytic internalization machinery predominates in the hyphal tip, forming a subapical collar. The spatial association of this collar with the apical region where secretory materials are delivered would allow removal of excess lipids/proteins reaching the plasma membrane with secretory vesicles (1, 2, 30, 49, 51, 57), but, most importantly, rapid endocytic recycling (i.e., efficient endocytosis of membrane proteins followed by their redelivery to the plasma membrane) can generate and maintain polarity, as shown with the v-SNARE and secretory-vesicle-resident SynA, which is a substrate of the subapical endocytic ring (1, 49, 52). It is plausible that such a mechanism could drive the polarization of one or more proteins acting as positional cues to mediate polarity maintenance.Genetic evidence strongly supported the conclusion that endocytosis is required for apical extension. Mutational inactivation of the A. nidulans fimbrin FimA or of the small GTPase ArfBArf6 (a regulator of endocytosis from fungi to mammals), led to delayed polarity establishment and morphologically aberrant tips indicative of polarity maintenance defects (30, 51). These mutations, although very severely debilitating, are not lethal. In contrast, heterokaryon rescue showed that SlaB, a key F-actin regulator of the endocytic internalization machinery (26), is essential in A. nidulans (2). slaBΔ cells are able to establish polarity, but they arrest in apical extension during the very early steps of polarity maintenance with a bud-like germ tube (2). However, work with Aspergillus oryzae using a thiamine-repressible promoter to drive A. oryzae End4 (AoEnd4) (SlaB) expression showed that although endocytosis was prevented and hyphal morphology altered under repressing conditions, hyphal tip extension and polarity maintenance were not completely abolished (20), perhaps suggesting that the phenotype of the thiamine-repressed conditional allele might not fully resemble the null phenotype.F-actin strongly predominates in the hyphal tips (2, 14, 17, 49, 51). Because endocytic internalization is powered by F-actin (27), predominance of endocytic “patches” (i.e., sites of endocytic internalization) in the tip accounts, at least in part, for F-actin polarization. However, F-actin plays fundamental nonendocytic roles in the tip; for example, actin cables nucleated by the SepA formin localizing to the apical crescent are thought to play a major role in secretion, whereas a network of F-actin microfilaments appears to be a major component of the Spitzenkörper (4, 21, 43, 49). Notably, all genes used to address the role of endocytosis in apical extension share in common their involvement in regulating F-actin. Thus, the Saccharomyces cerevisiae ArfB orthologue Arf3p regulates endocytosis but also appears to regulate F-actin at multiple levels (12, 28, 44). Like their respective S. cerevisiae orthologues Sla2p and Sac6p, SlaB and FimA are key components of endocytic patches, but in budding yeast their orthologues appear to regulate F-actin dynamics beyond endocytosis (27, 35, 56).To gain insight into the essential role of SlaB in A. nidulans, we designed a conditional expression allele. Time-lapse microscopy under restrictive conditions demonstrated that polarity establishment is essentially normal but that these mutant germ tubes arrested in apical extension subsequently undergo swelling, acquiring the characteristic bud-like shape of abortive slaBΔ germlings. Medium shift experiments allowed us to address the role of SlaB in apical extension beyond these early stages of polarity maintenance. We demonstrate the key role that SlaB plays in endocytosis and polarity maintenance, but we also show that deficiency of SlaB affects the actin cytoskeleton.  相似文献   

13.
Matrix effect is the alteration of an analyte''s concentration-signal response caused by co-existing ion components. With electrospray ionization (ESI), matrix effects are believed to be a function of the relative concentrations, ionization efficiency, and solvation energies of the analytes within the electrospray ionization droplet. For biological matrices such as plasma, the interactions between droplet components is immensely complex and the effect on analyte signal response not well elucidated. This study comprised of three sequential quantitative analyses: we investigated whether there is a generalizable correlation between the range of unique ions in a sample matrix (complexity); the amount of matrix components (concentration); and matrix effect, by comparing an E. coli digest matrix (∼2600 protein proteome) with phospholipid depleted human blood plasma, and unfractionated, nondepleted human plasma matrices (∼107 proteome) for six human plasma peptide multiple reaction monitoring assays. Our data set demonstrated analyte-specific interactions with matrix complexity and concentration properties resulting in significant ion suppression for all peptides (p < 0.01), with nonuniform effects on the ion signals of the analytes and their stable-isotope analogs. These matrix effects were then assessed for translation into relative residual error and precision effects in a low concentration (∼0–250 ng/ml) range across no-matrix, complex matrix, and highly complex matrix, when a standard addition stable isotope dilution calibration method was used. Relative residual error (%) and precision (CV%) by stable isotope dilution were within <20%; however, error in phospholipid-depleted and nondepleted plasma matrices were significantly higher compared with no-matrix (p = 0.006). Finally a novel reverse-polynomial dilution calibration method with and without phospholipid-depletion was compared with stable isotope dilution for relative residual error and precision. Reverse-polynomial dilution techniques extend the Lower Limit of Quantification and reduce error (p = 0.005) in low-concentration plasma peptide assays and is broadly applicable for verification phase Tier 2 multiplexed multiple reaction monitoring assay development within the FDA-National Cancer Institute (NCI) biomarker development pipeline.Plasma is the overriding human medium sampled for established and novel protein biomarkers (1, 2). As of 2011, 1929 high-confidence proteins have been cataloged by the Human Plasma Proteome Project, with estimates that there are up to 107 unique protein sequences in plasma that span a concentration range across 10 orders of magnitude (1, 3). 99% of the protein mass in plasma is made up of 22 proteins including Albumin, Fibrinogen, and a range of immunoglobulins, leaving more than 1900 known small proteins and essentially the entirety of the projected plasma proteome in the remaining 1% (4). It is these low-mass, low abundance proteins such as the Interleukins, C-Reactive Protein, and Carcinoma Antigen 125 (CA125), that are indicative of many important physiological and pathological processes, and proteomic scientists and clinicians have thus focused their efforts in qualitatively and quantitatively defining this fraction for novel biomarkers (46).The development of plasma biomarkers is a large-scale undertaking that spans discovery, verification, and validation phases in a multistage pipeline: Thousands of “discovered” differentiated proteins are evaluated for probability of effect, from which 10–100s of proteins are then selected for targeted quantification in verification phase to evaluate sensitivity and specificity for its intended indication (2, 7). Finally a panel of the strongest marker candidates is progressed to validation phase, and FDA-level validated quantitative assays are used to test the clinical utility of the biomarker panel. Liquid Chromatography coupled with Tandem Mass Spectrometry (LC-MS/MS)1 is the most robust analytical method available for proteomic scientists in this pipeline, able to separate complex mixtures and specifically and sensitively identify and quantify its components (2, 710), The ability to ionize and evaporate the contents of a liquid sample (coupling LC to MS/MS) is the basis that allows this to happen (9). Electrospray Ionization (ESI) is the most widely used ionization apparatus in LC-MS/MS bioanalysis because of its ionization efficiency and stability and low chemical specificity (9, 10). Although these properties make ESI very robust, the complexity of biological matrices poses a significant challenge for LC-ESI-MS/MS-based quantitation; despite chromatography and nanospray technology, the ESI droplet of a plasma peptide-digest sample (given its immense range of unique protein/peptide sequences and concentrations) can contain an unknown multitude of co-eluting components that “compete” to dissolve from the droplet and reach gas phase, suppressing and varying the signal intensity responses for a given analyte concentration (913). These ionization competing elements can also go on to produce isobaric signals in the third quadrupole that interfere with an analyte''s transition signals (14). Termed “matrix effects,” these phenomena of complex sample matrices can significantly impede quantitative accuracy (15). For high-throughput clinical assays, matrix effects are controlled for by preparing calibration standards in the same biological matrix to mimic the conditions of the samples intended for study as per FDA bioanalytical method validation guidelines (16). The catch to this technique is that the signal from the endogenous analyte in the background matrix hinders accuracy when the nominal concentration is close to or below the endogenous signal (14, 17). There is a need for broadly applicable methods of controlling matrix effects and increasing accuracy in low concentration MRM peptide assays for nondepleted, unfractionated plasma that can be adopted for the highly multiplexed, high throughput, “Tier 2” MS assays required in verification phase of the biomarker development pipeline (2, 8). Several simple methods have independently demonstrated the ability to increase accuracy in various hyphenated-MS assays in complex matrices: “Reverse” curves utilize the stable-isotope analog not as an internal standard but as a surrogate calibration analyte to circumvent interference from the endogenous analyte signal and extend assay Lower Limit(s) of Quantification (LLOQ), and nonlinear calibration techniques have proven to more accurately reflect the concentration-MS detector response at the low and high end of concentration gradients (8, 14, 1821). Specifically in the case of biological matrices, phospholipids are particularly deleterious ion suppressing elements because of their easily ionizable, polar, and hydrophobic moieties that can have complex interactions with co-eluting analytes as well as the chromatography stationary and mobile phases required for most other analytes (2225). Combination solid-phase extraction (SPE) and phospholipid removal techniques have proved to effectively minimize ion suppression effects in ESI-MS assays (2225).In this study, we investigated whether there is a generalizable linear correlation between the number of unique ions (complexity) in a biological sample matrix, the amount of ionizable matrix content (concentration), and matrix effects, for six human plasma peptides comparing serial dilutions of an Escherichia Coli (E. coli) peptide-digest against phospholipid-depleted and nondepleted unfractionated human plasma peptide-digest (highly complex) matrices. We examined the influence of matrix effects on relative residual error in a low-concentration (∼0–250 ng/ml) plasma peptide range, and compared the utility of a reverse-polynomial dilution (RPD) calibration method versus standard addition stable-isotope dilution (SID) in phospholipid-depleted and nondepleted unfractionated human plasma. A peptide-centric matrix effect is reported and the effect of the endogenous analyte signal on relative residual error in low-concentration (∼0–250 ng/ml) plasma peptide assays is established. A RPD calibration technique that extends LLOQ and reduces relative residual error in low-concentration plasma peptide MRM assays is presented.  相似文献   

14.
15.
Macrophages operate at the forefront of innate immunity and their discrimination of foreign versus “self” particles is critical for a number of responses including efficient pathogen killing, antigen presentation, and cytokine induction. In order to efficiently destroy the particles and detect potential threats, macrophages express an array of receptors to sense and phagocytose prey particles. In this study, we accurately quantified a proteomic time-course of isolated phagosomes from murine bone marrow-derived macrophages induced by particles conjugated to seven different ligands representing pathogen-associated molecular patterns, immune opsonins or apoptotic cell markers. We identified a clear functional differentiation over the three timepoints and detected subtle differences between certain ligand-phagosomes, indicating that triggering of receptors through a single ligand type has mild, but distinct, effects on phagosome proteome and function. Moreover, our data shows that uptake of phosphatidylserine-coated beads induces an active repression of NF-κB immune responses upon Toll-like receptor (TLR)-activation by recruitment of anti-inflammatory regulators to the phagosome. This data shows for the first time a systematic time-course analysis of bone marrow-derived macrophages phagosomes and how phagosome fate is regulated by the receptors triggered for phagocytosis.Macrophages exist in many different tissue subsets, are extremely plastic in response to cytokines and pathogen-associated molecular patterns and perform a wide range of biological functions (1, 2). One of the most important functions of macrophages is phagocytosis, defined as the active uptake of large particles (>0.5 μm) by cells (3). Phagocytosis is an important cellular mechanism for almost all eukaryotes, highly conserved in evolution (4), and, in mammals, is a key part of the innate immune response to invading microorganisms. Moreover, during homeostasis and development, macrophages phagocytose apoptotic cells and cell debris to recycle cellular building blocks (5, 6). Phagocytosis is induced through the binding of particles as diverse as microbes, apoptotic cells, or even inert beads to cell surface receptors. After internalization, newly formed phagosomes engage in a maturation process that involves fusion with various organelles, including endosomes and ultimately lysosomes (7, 8). This leads to the formation of phagolysosomes that degrade the foreign matter. Antigens from the particle are presented via MHC class I and II molecules, bridging innate and adaptive immunity.In order to effectively phagocytose the diverse types of particulates they can encounter, macrophages express a vast array of receptors to sense and respond to the different ligands; however, only a small subset are solely sufficient to trigger phagocytosis (9). The classic phagocytic receptors are the Fc receptor, which internalizes immunoglobulin-bound particles (10), and the complement receptors, which binds to complement-opsonized particles (11). Other well characterized ligands for phagocytic receptors include mannan, a polysaccharide common in bacterial membranes and fungal cell walls (12), that activates mannose receptors (13, 14); lipopolysaccharide (LPS)1, a glycolipid that constitutes the major portion of the outermost membrane of Gram-negative bacteria, that triggers CD14 as well as scavenger receptors and toll-like receptors (1520); and phosphatidylserine (PS), a lipid normally restricted to the inner leaflet of eukaryotic plasma membranes, but exposed in the outer leaflet during apoptosis. PS provides an “eat me” signal for macrophage clearance (21, 22) and triggers a range of receptors including TIM-4, BAI1, and Stabilin-2 (2327). Similarly, calreticulin, an endoplasmic reticulum protein that is also transported to the plasma membrane serves as a apoptotic signal has been proposed as a phagocytic ligand triggering the phagocytic receptor low-density lipoprotein receptor-related protein (LRP) (2830).Although it is established that phagosome function is affected by various activation states, including rate of maturation, degradative capacity, and antigen cross-presentation capabilities (3133), controversy exists around whether phagosome activity can be controlled directly, without prior activation, by receptor engagement at the phagosome level during biogenesis (3438). Here, we dissect the role that individual ligands play in controlling downstream phagosome maturation using a reductionist strategy of ligating single ligands to microparticles and analyzing resulting phagosomes by quantitative proteomics and fluorescent phagosome functional assays.  相似文献   

16.
17.
The Acanthamoeba castellanii myosin-Is were the first unconventional myosins to be discovered, and the myosin-I class has since been found to be one of the more diverse and abundant classes of the myosin superfamily. We used two-dimensional (2D) crystallization on phospholipid monolayers and negative stain electron microscopy to calculate a projection map of a “classical” myosin-I, Acanthamoeba myosin-IB (MIB), at ∼18 Å resolution. Interpretation of the projection map suggests that the MIB molecules sit upright on the membrane. We also used cryoelectron microscopy and helical image analysis to determine the three-dimensional structure of actin filaments decorated with unphosphorylated (inactive) MIB. The catalytic domain is similar to that of other myosins, whereas the large carboxy-terminal tail domain differs greatly from brush border myosin-I (BBM-I), another member of the myosin-I class. These differences may be relevant to the distinct cellular functions of these two types of myosin-I. The catalytic domain of MIB also attaches to F-actin at a significantly different angle, ∼10°, than BBM-I. Finally, there is evidence that the tails of adjacent MIB molecules interact in both the 2D crystal and in the decorated actin filaments.The myosin superfamily consists of at least 12 distinct classes that vary both in the sequence of their conserved myosin catalytic domains as well as in their unique tails (Mooseker and Cheney, 1995; Sellers and Goodson, 1995). For many years the only known myosins were the double-headed, filament-forming myosins found in muscle (conventional myosins or myosins-II). The remaining classes of myosin have been termed “unconventional myosins” to differentiate them from the myosins-II. Probably the most thoroughly studied class of unconventional myosins is the myosin-I class. These small, single-headed myosins bind to membrane lipids through a basic domain in their tail (for review see Pollard et al., 1991; Mooseker and Cheney, 1995). The first unconventional myosin (and first myosin-I) was isolated from Acanthamoeba castellanii (Pollard and Korn, 1973 a,b), and was purified on the basis of its K+, EDTA, and actin-activated MgATPase activities. However, this myosin was unusual in that it had a single heavy chain of ∼140 kD, in contrast to the two ∼200-kD heavy chains of myosin-II (Pollard and Korn, 1973 a).Three isoforms of the classical Acanthamoeba myosins-I are now known: myosins-IA, -IB, and -IC (Maruta and Korn, 1977a ,b; Maruta et al., 1979). Each of the isoforms consists of a conserved myosin catalytic domain, a binding site for one or two light chains, a basic domain, a GPA1(Q) domain (rich in glycine, proline and alanine [glutamine]), and an scr-homology domain-3 (SH3) domain (Pollard et al., 1991; Mooseker and Cheney, 1995). These myosins-I can associate with membranes or with actin filaments through their tail domains. An electrostatic association of myosin-I with anionic phospholipids and with base-stripped membranes has been shown to occur (Adams and Pollard, 1989; Miyata et al., 1989; Hayden et al., 1990), and this interaction has been mapped to the basic domain (Doberstein and Pollard, 1992). Interestingly, these myosins also contain a second, ATP-insensitive actin binding site (Lynch et al., 1986) enabling them to mediate actin–actin movements (Albanesi et al., 1985; Fujisaki et al., 1985). In myosin-IA (Lynch et al., 1986) and myosin-IC (Doberstein and Pollard, 1992), this binding site was localized to the GPA domain. Acanthamoeba myosins-I have maximal steady-state actin-activated ATPase rates of ∼10–20 s−1 (Pollard and Korn, 1973 b; Albanesi et al., 1983), and an unusual triphasic dependence upon actin concentration (Pollard and Korn, 1973 b; Albanesi et al., 1983). This triphasic activation is due to the actin cross-linking ability imparted by the ATP-insensitive actin binding site on the tail (Albanesi et al., 1985). Analysis of the individual steps in the ATPase cycle by transient kinetics revealed that the mechanism of myosin-IA is similar to slow skeletal muscle myosin, whereas myosin-IB (MIB) is similar to fast skeletal muscle myosin (Ostap and Pollard, 1996). The in vitro motility of myosin-I has also been well characterized (Zot et al., 1992). The maximal rate of filament sliding is ∼0.2 μm s−1. Interestingly, this rate is ∼10–50× slower than the rates observed for skeletal muscle myosin, even though the ATPase rates are comparable.MIB consists of a 125-kD heavy chain and a single 27-kD light chain (Maruta et al., 1979; Jung et al., 1987). This isoform is primarily associated with the plasma membrane as well as vacuolar membranes (Baines et al., 1992). MIB appears to be associated with the plasma membrane at sites of phagocytosis and was concentrated at the tips of pseudopodia (Baines et al., 1992). This localization suggests that MIB may be involved in membrane dynamics at the cell surface. MIB is regulated by heavy chain phosphorylation of serine 411 (Brzeska et al., 1989, 1990), which is located at the actin-binding site (Rayment et al., 1993). Similar to the myosin-I isoforms in Acanthamoeba, heavy chain phosphorylation results in >20-fold activation of the actin-activated myosin-I ATPase activity (Albanesi et al., 1983). This activation is not the result of changes in the binding of myosin-I to F-actin (Albanesi et al., 1983; Ostap and Pollard, 1996). The transient kinetic studies of Ostap and Pollard (1996) suggest that phosphorylation regulates the rate-limiting phosphate release step, the transition from weakly bound intermediates in rapid equilibrium with actin to strongly bound states, capable of sustaining force.Despite the extensive analysis of ameboid myosin-I biochemical properties and in vivo function, there is little structural information on these myosins. The only detailed structural information available for the myosins-I comes from recent electron microscopy studies on brush border myosin-I (BBM-I) (Jontes et al., 1995; Jontes and Milligan, 1997a ,b; Whittaker and Milligan, 1997), a structurally distinct myosin-I subtype. Therefore, we investigated the structure of a “classical,” ameboid-type myosin, Acanthamoeba MIB using electron microscopy. First, electron micrographs of negatively stained two-dimensional (2D) crystals were used to generate a projection map of MIB at ∼18 Å resolution. In addition, we used cryoelectron microscopy and helical image analysis to produce a moderate resolution three-dimensional (3D) map (30 Å) of actin filaments decorated with MIB. These studies enabled us to compare the structure of MIB with BBM-I. The comparison of MIB with BBM-I reveals marked structural differences in the tail domains of these two proteins; MIB appears to have a much shorter “lever arm” and a more compact tail, whereas most of the BBM-I mass is composed of an extended light chain–binding domain (LCBD). In addition, the MIB catalytic domain appears to be slightly tilted compared to BBM-I, with respect to the F-actin axis. Our structural results suggest that these two types of myosin-I may have distinct intracellular functions.  相似文献   

18.
Significant progress in instrumentation and sample preparation approaches have recently expanded the potential of MALDI imaging mass spectrometry to the analysis of phospholipids and other endogenous metabolites naturally occurring in tissue specimens. Here we explore some of the requirements necessary for the successful analysis and imaging of phospholipids from thin tissue sections of various dimensions by MALDI time-of-flight mass spectrometry. We address methodology issues relative to the imaging of whole-body sections such as those cut from model laboratory animals, sections of intermediate dimensions typically prepared from individual organs, as well as the requirements for imaging areas of interests from these sections at a cellular scale spatial resolution. We also review existing limitations of MALDI imaging MS technology relative to compound identification. Finally, we conclude with a perspective on important issues relative to data exploitation and management that need to be solved to maximize biological understanding of the tissue specimen investigated.Since its introduction in the late 90s (1), MALDI imaging mass spectrometry (MS) technology has witnessed a phenomenal expansion. Initially introduced for the mapping of intact proteins from fresh frozen tissue sections (2), imaging MS is now routinely applied to a wide range of different compounds including peptides, proteins, lipids, metabolites, and xenobiotics (37). Numerous compound-specific sample preparation protocols and analytical strategies have been developed. These include tissue sectioning and handling (814), automated matrix deposition approaches and data acquisition strategies (1521), and the emergence of in situ tissue chemistries (2225). Originally performed on sections cut from fresh frozen tissue specimens, methodologies incorporating an in situ enzymatic digestion step prior to matrix application have been optimized to access the proteome locked in formalin-fixed paraffin-embedded tissue biopsies (2529). The possibility to use tissues preserved using non-cross-linking approaches has also been demonstrated (3032). These methodologies are of high importance for the study of numerous diseases because they potentially allow the retrospective analysis for biomarker validation and discovery of the millions of tissue biopsies currently stored worldwide in tissue banks and repositories.In the past decade, instrumentation for imaging MS has also greatly evolved. Whereas the first MS images were collected with time-of-flight instruments (TOF) capable of repetition rates of a few hertz, modern systems are today capable of acquiring data in the kilohertz range and above with improved sensitivity, mass resolving power, and accuracy, significantly reducing acquisition time and improving image quality (33, 34). Beyond time-of-flight analyzers, other MALDI-based instruments have been used such as ion traps (3537), Qq TOF instruments (3840), and trap-TOF (16, 41). Ion mobility technology has also been used in conjunction with imaging MS (4244). More recently, MALDI FT/ICR and Orbitrap mass spectrometers have been demonstrated to be extremely valuable instruments for the performance of imaging MS at very high mass resolving power (4547). These non-TOF-based systems have proven to be extremely powerful for the imaging of lower molecular weight compounds such as lipids, drugs, and metabolites. Home-built instrumentation and analytical approaches to probe tissues at higher spatial resolution (1–10 μm) have also been described (4850). In parallel to instrumentation developments, automated data acquisition, image visualization, and processing software packages have now also been developed by most manufacturers.To date, a wide range of biological systems have been studied using imaging MS as a primary methodology. Of strong interest are the organization and identification of the molecular composition of diseased tissues in direct correlation with the underlying histology and how it differs from healthy tissues. Such an approach has been used for the study of cancers (5154), neurologic disorders (5557), and other diseases (58, 59). The clinical potential of the imaging MS technology is enormous (7, 60, 61). Results give insights into the onset and progression of diseases, identify novel sets of disease-specific markers, and can provide a molecular confirmation of diagnosis as well as aide in outcome prediction (6264). Imaging MS has also been extensively used to study the development, functioning, and aging of different organs such as the kidney, prostate, epididymis, and eye lens (6570). Beyond the study of isolated tissues or organs, whole-body sections from several model animals such as leeches, mice, and rats have been investigated (7174). For these analyses, specialized instrumentation and protocols are necessary for tissue sectioning and handling (72, 73). Whole-body imaging MS opens the door to the study of the localization and accumulation of administered pharmaceuticals and their known metabolites at the level of entire organisms as well as the monitoring of their efficacy or toxicity as a function of time or dose (72, 73, 75, 76).There is considerable interest in determining the identification and localization of small biomolecules such as lipids in tissues because they are involved in many essential biological functions including cell signaling, energy storage, and membrane structure and function. Defects in lipid metabolism play a role in many diseases such as muscular dystrophy and cardiovascular disease. Phospholipids in tissues have been intensively studied by several groups (37, 40, 7783). In this respect, for optimal recovery of signal, several variables such as the choice of matrix for both imaging and fragmentation, solvent system, and instrument polarity have been investigated (20, 84). Particularly, the use of lithium cation adducts to facilitate phospholipid identification by tandem MS directly from tissue has also been reported (85). Of significant interest is the recent emergence of two new solvent-free matrix deposition approaches that perform exceptionally well for phospholipid imaging analyses. The first approach, described by Hankin et al. (86), consists in depositing the matrix on the sections through a sublimation process. The described sublimation system consists of sublimation glassware, a heated sand or oil bath (100–200 °C), and a primary vacuum pump (∼5 × 10−2 torr). Within a few minutes of initiating the sublimation process, an exceptionally homogeneous film of matrix forms on the section. The thickness of the matrix may be controlled by regulating pressure, temperature, and sublimation time. The second approach, described by Puolitaival et al.(87), uses a fine mesh sieve (≤20 μm) to filter finely ground matrix on the tissue sections. Agitation of the sieve results in passage of the matrix through the mesh and the deposition of a fairly homogeneous layer of submicrometer matrix crystals of the surface of the sections. The matrix density on the sections is controlled by direct observation using a standard light microscope. This matrix deposition approach was also found to be ideal to image certain drug compounds (88, 89). Both strategies allow very rapid production of homogeneous matrix coatings on tissue sections with a fairly inexpensive setup. Signal recovery was found to be comparable with those obtained by conventional spray deposition. With the appropriate size sublimation device or sieve, larger sections with dimensions of several centimeters such as those cut from mouse or rat whole bodies can also be rapidly and homogeneously coated.Here we present several examples of MALDI imaging MS of phospholipids from tissue sections using TOF mass spectrometers over a wide range of dimensions from whole-body sections (several centimeters), to individual organs (several millimeters), down to high spatial resolution imaging of selected tissue areas (hundreds of micrometers) at 10-μm lateral resolution and below. For all of these dimension ranges, technological considerations and practical aspects are discussed. In light of the imaging MS results, we also address issues faced for compound identification by tandem MS analysis performed directly on the sections. Finally, we discuss under “Perspective” our vision of the future of the field as well as the technological improvements and analytical tools that need to be improved upon and developed.  相似文献   

19.
Bone samples from several vertebrates were collected from the Ziegler Reservoir fossil site, in Snowmass Village, Colorado, and processed for proteomics analysis. The specimens come from Pleistocene megafauna Bison latifrons, dating back ∼120,000 years. Proteomics analysis using a simplified sample preparation procedure and tandem mass spectrometry (MS/MS) was applied to obtain protein identifications. Several bioinformatics resources were used to obtain peptide identifications based on sequence homology to extant species with annotated genomes. With the exception of soil sample controls, all samples resulted in confident peptide identifications that mapped to type I collagen. In addition, we analyzed a specimen from the extinct B. latifrons that yielded peptide identifications mapping to over 33 bovine proteins. Our analysis resulted in extensive fibrillar collagen sequence coverage, including the identification of posttranslational modifications. Hydroxylysine glucosylgalactosylation, a modification thought to be involved in collagen fiber formation and bone mineralization, was identified for the first time in an ancient protein dataset. Meta-analysis of data from other studies indicates that this modification may be common in well-preserved prehistoric samples. Additional peptide sequences from extracellular matrix (ECM) and non-ECM proteins have also been identified for the first time in ancient tissue samples. These data provide a framework for analyzing ancient protein signatures in well-preserved fossil specimens, while also contributing novel insights into the molecular basis of organic matter preservation. As such, this analysis has unearthed common posttranslational modifications of collagen that may assist in its preservation over time. The data are available via ProteomeXchange with identifier PXD001827.During the last decade, paleontology and taphonomy (the study of decaying organisms over time and the fossilization processes) have begun to overlap with the field of proteomics to shed new light on preserved organic matter in fossilized bones (14). These bones represent a time capsule of ancient biomolecules, owing to their natural resistance to post mortem decay arising from a unique combination of mechanical, structural, and chemical properties (47).Although bones can be cursorily described as a composite of collagen (protein) and hydroxyapatite (mineral), fossilized bones undergo three distinct diagenesis pathways: (i) chemical deterioration of the organic phase; (ii) chemical deterioration of the mineral phase; and (iii) (micro)biological attack of the composite (6). In addition, the rate of these degradation pathways are affected by temperature, as higher burial temperatures have been shown to accelerate these processes (6, 8). Though relatively unusual, the first of these three pathways results in a slower deterioration process, which is more generally mitigated under (6) specific environmental constraints, such as geochemical stability (stable temperature and acidity) that promote bone mineral preservation. Importantly, slower deterioration results in more preserved biological materials that are more amenable to downstream analytical assays. One example of this is the controversial case of bone and soft-tissue preservation from the Cretaceous/Tertiary boundary (922). In light of these and other studies of ancient biomolecules, paleontological models have proposed that organic biomolecules in ancient samples, such as collagen sequences from the 80 million-year-(my)-old Campanian hadrosaur, Brachylophosaurus canadensis (16) or 68-my-old Tyrannosaurus rex, might be protected by the microenvironment within bones. Such spaces are believed to form a protective shelter that is able to reduce the effects of diagenetic events. In addition to collagen, preserved biomolecules include blood proteins, cellular lipids, and DNA (4, 5). While the maximum estimated lifespan of DNA in bones is ∼20,000 years (ky) at 10 °C, bone proteins have an even longer lifespan, making them an exceptional target for analysis to gain relevant insights into fossilized samples (6). Indeed, the survival of collagen, which is considered to be the most abundant bone protein, is estimated to be in the range 340 ky at 20 °C. Similarly, osteocalcin, the second-most abundant bone protein, can persist for ≈45 ky at 20 °C, thus opening an unprecedented analytical window to study extremely old samples (2, 4, 23).Although ancient DNA amplification and sequencing can yield interesting clues and potential artifacts from contaminating agents (7, 2428), the improved preservation of ancient proteins provides access to a reservoir of otherwise unavailable genetic information for phylogenetic inference (25, 29, 30). In particular, mass spectrometry (MS)-based screening of species-specific collagen peptides has recently been used as a low-cost, rapid alternative to DNA sequencing for taxonomic attribution of morphologically unidentifiable small bone fragments and teeth stemming from diverse archeological contexts (25, 3133).For over five decades, researchers have presented biochemical evidence for the existence of preserved protein material from ancient bone samples (3436). One of the first direct measurements was by amino acid analysis, which showed that the compositional profile of ancient samples was consistent with collagens in modern bone samples (3739). Preservation of organic biomolecules, either from bone, dentin, antlers, or ivory, has been investigated by radiolabeled 14C fossil dating (40) to provide an avenue of delineating evolutionary divergence from extant species (3, 41, 42). It is also important to note that these parameters primarily depend on ancient bone collagen as the levels remain largely unchanged (a high percentage of collagen is retained, as gleaned by laboratory experiments on bone taphonomy (6)). Additionally, antibody-based immunostaining methods have given indirect evidence of intact peptide amide bonds (4345) to aid some of the first evidence of protein other than collagen and osteocalcin in ancient mammoth (43) and human specimens (46).In the past, mass spectrometry has been used to obtain MS signals consistent with modern osteocalcin samples (2, 47), and eventually postsource decay peptide fragmentation was used to confirm the identification of osteocalcin in fossil hominids dating back ∼75 ky (48). More recently, modern “bottom-up” proteomic methods were applied to mastodon and T. rex samples (10), complementing immunohistochemistry evidence (13, 17). The results hinted at the potential of identifying peptides from proteolytic digest of well-preserved bone samples. This work also highlighted the importance of minimizing sources of protein contamination and adhering to data publication guidelines (20, 21). In the past few years, a very well-preserved juvenile mammoth referred to as Lyuba was discovered in the Siberian permafrost and analyzed using high-resolution tandem mass spectrometry (29). This study was followed with a report by Wadsworth and Buckley (30) describing the analysis of proteins from 19 bovine bone samples spanning 4 ky to 1.5 my. Both of these groups reported the identification of additional collagen and noncollagen proteins.Recently, a series of large extinct mammal bones were unearthed at a reservoir near Snowmass Village, Colorado, USA (49, 50). The finding was made during a construction project at the Ziegler Reservoir, a fossil site that was originally a lake formed at an elevation of ∼2,705 m during the Bull Lake glaciations ∼140 ky ago (49, 51). The original lake area was ∼5 hectares in size with a total catchment of ∼14 hectares and lacked a direct water flow inlet or outlet. This closed drainage basin established a relatively unique environment that resulted in the exceptional preservation of plant material, insects (52), and vertebrate bones (49). In particular, a cranial specimen from extinct Bison latifrons was unearthed from the Biostratigraphic Zone/Marine Oxygen Isotope Stage (MIS) 5d, which dates back to ∼120 ky (53, 54).Here, we describe the use of paleoproteomics, for the identification of protein remnants with a focus on a particularly unique B. latifrons cranial specimen found at the Ziegler site. We developed a simplified sample processing approach that allows for analysis of low milligram quantities of ancient samples for peptide identification. Our method avoids the extensive demineralization steps of traditional protocols and utilizes an acid labile detergent to allow for efficient extraction and digestion without the need for additional sample cleanup steps. This approach was applied to a specimen from B. latifrons that displayed visual and mechanical properties consistent with the meninges, a fibrous tissue that lines the cranial cavity. Bioinformatics analysis revealed the presence of a recurring glycosylation signature in well-preserved collagens. In particular, the presence of glycosylated hydroxylysine residues was identified as a unique feature of bone fossil collagen, as gleaned through meta-analyses of raw data from previous reports on woolly mammoth (Mammuthus primigenius) and bovine samples (29, 30). The results from these meta-analyses indicate a common, unique feature of collagen that coincides with, and possibly contributes to its preservation.  相似文献   

20.
Current analytical strategies for collecting proteomic data using data-dependent acquisition (DDA) are limited by the low analytical reproducibility of the method. Proteomic discovery efforts that exploit the benefits of DDA, such as providing peptide sequence information, but that enable improved analytical reproducibility, represent an ideal scenario for maximizing measureable peptide identifications in “shotgun”-type proteomic studies. Therefore, we propose an analytical workflow combining DDA with retention time aligned extracted ion chromatogram (XIC) areas obtained from high mass accuracy MS1 data acquired in parallel. We applied this workflow to the analyses of sample matrixes prepared from mouse blood plasma and brain tissues and observed increases in peptide detection of up to 30.5% due to the comparison of peptide MS1 XIC areas following retention time alignment of co-identified peptides. Furthermore, we show that the approach is quantitative using peptide standards diluted into a complex matrix. These data revealed that peptide MS1 XIC areas provide linear response of over three orders of magnitude down to low femtomole (fmol) levels. These findings argue that augmenting “shotgun” proteomic workflows with retention time alignment of peptide identifications and comparative analyses of corresponding peptide MS1 XIC areas improve the analytical performance of global proteomic discovery methods using DDA.Label-free methods in mass spectrometry-based proteomics, such as those used in common “shotgun” proteomic studies, provide peptide sequence information as well as relative measurements of peptide abundance (13). A common data acquisition strategy is based on data-dependent acquisition (DDA)1 where the most abundant precursor ions are selected for tandem mass spectrometry (MS/MS) analysis (12). DDA attempts to minimize redundant peptide precursor selection and maximize the depth of proteome coverage (2). However, the analytical reproducibility of peptide identifications obtained using DDA-based methods result in <75% overlap between technical replicates (34). Comparisons of peptide identifications between replicate analyses have shown that the rate of new peptide identifications increases sharply following two replicate sample injections and gradually tapers off after approximately five replicate injections (4). This phenomenon is due, in part, to the semirandom sampling of peptides in a DDA experiment (5).Alternate label-free methods focused on measuring the abundance of intact peptide ions, such as the accurate mass and time tag (AMT) approach (68, 42), are aimed at differential analyses of extracted ion chromatogram (XIC) areas integrated from high mass accuracy peptide precursor mass spectra (MS1 spectra) exhibiting discrete chromatographic elution times. This method is particularly powerful for the analysis of post-translationally modified (PTM) peptides as pairing the low abundance of PTM candidates with the variable nature of DDA complicates comparisons between samples (9, 43). However, label-free strategies focused on the analysis of peptide MS1 XIC areas are dependent on a priori knowledge of peptide ions and retention times (210). Thus, prospective analyses of samples are needed to assess peptides and their respective retention times. This prospective analysis may not be possible for reagent-limited samples. Further, the usage of previously established peptide features in the analysis of different sample types can be confounded by unknown matrix effects that can produce variable retention time characteristics and peptide ion suppression (2). Therefore, proteomic strategies that make use of DDA, to provide peptide sequence information and identify features within the sample, but that also use MS1 data for comparisons between samples, represent an ideal combination for maximizing measureable peptide identification events in “shotgun” proteomic discovery analyses.Here we describe an analytical workflow that combines traditional DDA methods with the analysis of retention time aligned XIC areas extracted from high mass accuracy peptide precursor MS1 spectra. This method resulted in a 25.1% (±6.6%) increase in measureable peptide identification events across samples of diverse composition because of the inferential extraction of peptide MS1 XIC areas in sample sets lacking corresponding MS/MS events. These findings were observed in measurements of peptide MS1 XIC abundances using sample types ranging from tryptic digests of olfactory bulb tissues dissected from Homer2 knockout and wild-type mice to mouse blood plasma exhibiting differential levels of hemolysis. We further establish that this method is quantitative using a dilution series of known bovine standard peptide concentrations spiked into mouse blood plasma. These data show that comparative analysis between samples should be performed using peptide MS1 data as opposed to semirandomly sampled peptide MS/MS data. This approach maximizes the number of peptides that can be compared between samples.  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