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 78 毫秒
1.
The dengue fever epidemic in Guangzhou may have been affected by the Coronavirus Disease 2019 (COVID-19) pandemic. The number of dengue cases dropped drastically in 2020, and there have been only 2 local cases, suggesting that dengue has not become endemic in Guangzhou.

Guangzhou is located on the southeast coast of China and is the country’s third largest city. Since 1978, outbreaks of dengue fever have occurred intermittently in this city. In the past decade, the number of reported dengue cases reached more than 1,000 in 2013, 2014, 2018, and 2019, with 37,385 cases reported in 2014 alone. Therefore, dengue fever is a major public health concern in Guangzhou, and there is a continuing argument that it is endemic in Guangzhou [13].The numbers of dengue cases from 2017 to 2020 are shown in Table 1. In 2020, the total and local case numbers dropped dramatically compared to the previous years. With a high proportion of imported cases (n = 32, 94.12%), the proportion of local cases (n = 2, 5.88%) was considerably low in 2020. All the prevention and control strategies for dengue, including issuing public education messages, preventing further mosquito bites in patients, cleaning vector breeding sites, and using pesticides, were similar during these years. Additionally, dengue, as a mosquito-borne viral infectious disease, is closely related to mosquito density. The mosquito ovitrap index (MOI), which is the proportion of positive mosquito ovitraps, is usually used to indicate mosquito density. The MOI in 2017, 2018, and 2019 was 7.073 ± 1.016, 9.657 ± 1.307, and 8.464 ± 0.961, respectively. The average MOI was 8.398 ± 0.648 from 2017 to 2019 in Guangzhou. The MOI in 2020 was 7.135 ± 0.786, which remained at the median risk level. Therefore, the abnormal decline in dengue cases could not be attributed to the change in mosquito density in 2020.Table 1Numbers and percentages of dengue cases from 2017 to 2019.
Year2017201820192020
Total cases9441,2951,65534
Imported cases(percentage)69 (7.31%)96 (7.41%)270 (16.31%)32 (94.12%)
Local cases (percentage)875 (92.69%)1,199 (92.59%)1,385 (83.69%)2 (5.88%)
Open in a separate windowIn 2020, the 14-day quarantine in a designated hotel for international travelers to curb the spread of Coronavirus Disease 2019 (COVID-19) was an important public health intervention. People had to remain indoors except for medical care needs. All imported dengue cases were identified during their quarantine periods. No secondary case related to the imported cases was reported. This may be because Aedes albopictus, which is the major vector of dengue in Guangzhou, bites aggressively during the day outdoors. The chance of being bitten by A. albopictus was reduced by staying all day indoors. Moreover, some research revealed that viremia occurred 6 to 18 hours before symptoms appeared and lasted as long as 12 days [4]. After the 14-day quarantine, viremia had almost subsided. Therefore, imported dengue cases were unlikely to be transmitted. The impact of imported dengue cases was limited by the quarantine, which provided a rare opportunity to identify the local epidemic.The epidemiology investigation showed that the 2 local cases, who were living in the same building, had no travel history outside Guangzhou in 2020 and had symptoms successively. Two dengue virus serotype 2 (DENV-2) strains were isolated from them. The envelope gene sequences were obtained and deposited in GenBank under accession numbers MW295818 and MW345921. Reference sequences, which were downloaded from GenBank, and sequences of Guangzhou strains identified in the previous years, were used to construct a phylogenetic tree. The 2 isolated strains in 2020 were identical. The tree (Fig 1) shows that the 2 strains belonged to the Malaysia/Indian subcontinent genotype, which was the prevailing genotype in Guangzhou [5]. However, they were neither identical with nor derived from the Guangzhou strains obtained from the previous years. Using the Basic Local Alignment Search Tool in GenBank, the 2 strains were found to be highly similar to those identified in Zhejiang (China), Singapore, and Guangdong (China) in 2017. These results imply that the local cases may be secondary to some undiscovered cases imported from other cities in China, as no restriction and quarantine was imposed for domestic travels.Open in a separate windowFig 1Maximum-likelihood phylogenetic tree shows the evolutionary relationships of DENV-2 detected in the sera of 2 local cases along with 45 other sequences.The reference sequences are named using the GenBank accession number, country, and year. The sequences of strains isolated in Guangzhou are named using the GenBank accession number, year, and our lab number. Bootstrap support values are shown in the notes. Strains isolated in 2020 are indicated with a black triangle.When the impact of imported dengue cases was limited by quarantine, dengue did not spread in Guangzhou during 2020, with the MOI still at median risk level and without any changes in the prevention and control strategies. Moreover, serotype 1 had been prevalent in Guangzhou since 2011 [6,7]. However, there was no local infection of serotype 1 detected in 2020. These observations may provide further evidence that dengue fever is not endemic in Guangzhou.In conclusion, the number of dengue cases decreased during the COVID-19 epidemic in Guangzhou in 2020. Thus, we believe that dengue fever is not endemic in Guangzhou.  相似文献   

2.
Seedling roots enable plant establishment. Their small phenotypes are measured routinely. Adult root systems are relevant to yield and efficiency, but phenotyping is challenging. Root length exceeds the volume of most pots. Field studies measure partial adult root systems through coring or use seedling roots as adult surrogates. Here, we phenotyped 79 diverse lines of the small grass model Brachypodium distachyon to adults in 50-cm-long tubes of soil with irrigation; a subset of 16 lines was droughted. Variation was large (total biomass, ×8; total root length [TRL], ×10; and root mass ratio, ×6), repeatable, and attributable to genetic factors (heritabilities ranged from approximately 50% for root growth to 82% for partitioning phenotypes). Lines were dissected into seed-borne tissues (stem and primary seminal axile roots) and stem-borne tissues (tillers and coleoptile and leaf node axile roots) plus branch roots. All lines developed one seminal root that varied, with branch roots, from 31% to 90% of TRL in the well-watered condition. With drought, 100% of TRL was seminal, regardless of line because nodal roots were almost always inhibited in drying topsoil. Irrigation stimulated nodal roots depending on genotype. Shoot size and tillers correlated positively with roots with irrigation, but partitioning depended on genotype and was plastic with drought. Adult root systems of B. distachyon have genetic variation to exploit to increase cereal yields through genes associated with partitioning among roots and their responsiveness to irrigation. Whole-plant phenotypes could enhance gain for droughted environments because root and shoot traits are coselected.Adult plant root systems are relevant to the size and efficiency of seed yield. They supply water and nutrients for the plant to acquire biomass, which is positively correlated to the harvest index (allocation to seed grain), and the stages of flowering and grain development. Modeling in wheat (Triticum aestivum) suggested that an extra 10 mm of water absorbed by such adult root systems during grain filling resulted in an increase of approximately 500 kg grain ha−1 (Manschadi et al., 2006). This was 25% above the average annual yield of wheat in rain-fed environments of Australia. This number was remarkably close to experimental data obtained in the field in Australia (Kirkegaard et al., 2007). Together, these modeling and field experiments have shown that adult root systems are critical for water absorption and grain yield in cereals, such as wheat, emphasizing the importance of characterizing adult root systems to identify phenotypes for productivity improvements.Most root phenotypes, however, have been described for seedling roots. Seedling roots are essential for plant establishment, and hence, the plant’s potential to set seed. For technical reasons, seedlings are more often screened than adult plants because of the ease of handling smaller plants and the high throughput. Seedling-stage phenotyping may also improve overall reproducibility of results because often, growth media are soil free. Seedling soil-free root phenotyping conditions are well suited to dissecting fine and sensitive mechanisms, such as lateral root initiation (Casimiro et al., 2003; Péret et al., 2009a, 2009b). A number of genes underlying root processes have been identified or characterized using seedlings, notably with the dicotyledonous models Arabidopsis (Arabidopsis thaliana; Mouchel et al., 2004; Fitz Gerald et al., 2006; Yokawa et al., 2013) and Medicago truncatula (Laffont et al., 2010) and the cereals maize (Zea mays; Hochholdinger et al., 2001) and rice (Oryza sativa; Inukai et al., 2005; Kitomi et al., 2008).Extrapolation from seedling to adult root systems presents major questions (Hochholdinger and Zimmermann, 2008; Chochois et al., 2012; Rich and Watt, 2013). Are phenotypes in seedling roots present in adult roots given developmental events associated with aging? Is expression of phenotypes correlated in seedling and adult roots if time compounds effects of growth rates and growth conditions on roots? Watt et al. (2013) showed in wheat seedlings that root traits in the laboratory and field correlated positively but that neither correlated with adult root traits in the field. Factors between seedling and adult roots seemed to be differences in developmental stage and the time that growing roots experience the environment.Seedling and adult root differences may be larger in grasses than dicotyledons. Grass root systems have two developmental components: seed-borne (seminal) roots, of which a number emerge at germination and continue to grow and branch throughout the plant life, and stem-borne (nodal or adventitious) roots, which emerge from around the three-leaf stage and continue to emerge, grow, and branch throughout the plant life. Phenotypes and traits of adult root systems of grasses, which include the major cereal crops wheat, rice, and maize, are difficult to predict in seedling screens and ideally identified from adult root systems first (Gamuyao et al., 2012).Phenotyping of adult roots is possible in the field using trenches (Maeght et al., 2013) or coring (Wasson et al., 2014). A portion of the root system is captured with these methods. Alternatively, entire adult root systems can be contained within pots dug into the ground before sowing. These need to be large; field wheat roots, for example, can reach depths greater than 1.5 m depending on genotype and environment. This method prevents root-root interactions that occur under normal field sowing of a plant canopy and is also a compromise.A solution to the problem of phenotyping adult cereal root systems is a model for monocotyledon grasses: Brachypodium distachyon. B. distachyon is a small-stature grass with a small genome that is fully sequenced (Vogel et al., 2010). It has molecular tools equivalent to those available in Arabidopsis (Draper et al., 2001; Brkljacic et al., 2011; Mur et al., 2011). The root system of B. distachyon reference line Bd21 is more similar to wheat than other model and crop grasses (Watt et al., 2009). It has a seed-borne primary seminal root (PSR) that emerges from the embryo at seed germination and multiple stem-borne coleoptile node axile roots (CNRs) and leaf node axile roots (LNRs), also known as crown roots or adventitious roots, that emerge at about three leaves through to grain development. Branch roots emerge from all root types. There are no known anatomical differences between root types of wheat and B. distachyon (Watt et al., 2009). In a recent study, we report postflowering root growth in B. distachyon line Bd21-3, showing that this model can be used to answer questions relevant to the adult root systems of grasses (Chochois et al., 2012).In this study, we used B. distachyon to identify adult plant phenotypes related to the partitioning among seed-borne and stem-borne shoots and roots for the genetic improvement of well-watered and droughted cereals (Fig. 1; Krassovsky, 1926; Navara et al., 1994), nitrogen, phosphorus (Tennant, 1976; Brady et al., 1995), oxygen (Wiengweera and Greenway, 2004), soil hardness (Acuna et al., 2007), and microorganisms (Sivasithamparam et al., 1978). Of note is the study by Krassovsky (1926), which was the first, to our knowledge, to show differences in function related to water. Krassovsky (1926) showed that seminal roots of wheat absorbed almost 2 times the water as nodal roots per unit dry weight but that nodal roots absorbed a more diluted nutrient solution than seminal roots. Krassovsky (1926) also showed by removing seminal or nodal roots as they emerged that “seminal roots serve the main stem, while nodal roots serve the tillers” (Krassovsky, 1926). Volkmar (1997) showed, more recently, in wheat that nodal and seminal roots may sense and respond to drought differently. In millet (Pennisetum glaucum) and sorghum (Sorghum bicolor), Rostamza et al. (2013) found that millet was able to grow nodal roots in a dryer soil than sorghum, possibly because of shoot and root vigor.Open in a separate windowFigure 1.B. distachyon plant scanned at the fourth leaf stage, with the root and shoot phenotypes studied indicated. Supplemental Table S1.
PhenotypeAbbreviationUnitRange of Variation
All Experiments (79 Lines and 582 Plants)Experiment 6 (36 Lines)
Whole plant
TDWTDWMilligrams88.6–773.8 (×8.7)285.6–438 (×1.5)
Shoot
SDWSDWMilligrams56.4–442.5 (×7.8)78.2–442.5 (×5.7)
 No. of tillersTillerNCount2.8–20.3 (×7.4)10–20.3 (×2)
Total root system
TRLTRLCentimeters1,050–10,770 (×10.3)2,090–5,140 (×2.5)
RDWRDWMilligrams28.9–312.17 (×10.8)62.2–179.1 (×2.9)
RootpcRootpcPercentage (of TDW)20.5–60.6 (×3)20.5–44.3 (×2.2)
R/SR/SUnitless ratio0.26–1.54 (×6)0.26–0.80 (×3.1)
PSRs
 Length (including branch roots)PSRLCentimeters549.1–4,024.6 (×7.3)716–2,984 (×4.2)
PSRpcPSRpcPercentage (of TRL)14.9–94.1 (×6.3)31.3–72.3 (×2.3)
 No. of axile rootsPSRcountCount11
 Length of axile rootPSRsumCentimeters17.45–52 (×3)17.45–30.3 (×1.7)
 Branch rootsPSRbranchCentimeters · (centimeters of axile root)−119.9–109.3 (×5.5)29.3–104.3 (×3.6)
CNRs
 Length (including branch roots)CNRLCentimeters0–3,856.70–2,266.5
CNRpcCNRpcPercentage (of TRL)0–57.10–49.8
 No. of axile rootsCNRcountCount0–20–2
 Cumulated length of axile rootsCNRsumCentimeters0–113.90–47.87
 Branch rootsCNRbranchCentimeters · (centimeters of axile root)−10–77.80–77.8
LNRs
 Length (including branch roots)LNRLCentimeters99.5–5,806.5 (×58.5)216.1–2,532.4 (×11.7)
LNRpcLNRpcPercentage (of TRL)4.2–72.7 (×17.5)6–64.8 (×10.9)
LNRcountLNRcountCount2–22.2 (×11.1)3.3–15.3 (×4.6)
LNRsumLNRsumCentimeters25.9–485.548–232 (×4.8)
 Branch rootsLNRbranchCentimeters · (centimeters of axile root)−12.1–25.4 (×12.1)3.2–15.9 (×5)
Open in a separate windowThe third reason for dissecting the different root types in this study was that they seem to have independent genetic regulation through major genes. Genes affecting specifically nodal root growth have been identified in maize (Hetz et al., 1996; Hochholdinger and Feix, 1998) and rice (Inukai et al., 2001, 2005; Liu et al., 2005, 2009; Zhao et al., 2009; Coudert et al., 2010; Gamuyao et al., 2012). Here, we also dissect branch (lateral) development on the seminal or nodal roots. Genes specific to branch roots have been identified in Arabidopsis (Casimiro et al., 2003; Péret et al., 2009a), rice (Hao and Ichii, 1999; Wang et al., 2006; Zheng et al., 2013), and maize (Hochholdinger and Feix, 1998; Hochholdinger et al., 2001; Woll et al., 2005).This study explored the hypothesis that adult root systems of B. distachyon contain genotypic variation that can be exploited through phenotyping and genotyping to increase cereal yields. A selection of 79 wild lines of B. distachyon from various parts of the Middle East (Fig. 2 shows the geographic origins of the lines) was phenotyped. They were selected for maximum genotypic diversity from 187 diploid lines analyzed with 43 simple sequence repeat markers (Vogel et al., 2009). We phenotyped shoots and mature root systems concurrently because B. distachyon is small enough to complete its life cycle in relatively small pots of soil with minimal influence of pot size compared with crops, such as wheat. We further phenotyped a subset of this population under irrigation (well watered) and drought to assess genotype response to water supply. By conducting whole-plant studies, we aimed to identify phenotypes that described partitioning among shoot and root components and within seed-borne and stem-borne roots. Phenotypes that have the potential to be beneficial to shoot and root components may speed up genetic gain in future.Open in a separate windowFigure 2.B. distachyon lines phenotyped in this study and their geographical origin. Capital letters in parentheses indicate the country of origin: Turkey (T), Spain (S), and Iraq (I; Vogel et al., 2009). a, Adi3, Adi7, Adi10, Adi12, Adi13, and Adi15; b, Bd21 and Bd21-3 are the reference lines of this study. Bd21 was the first sequenced line (Vogel et al., 2010) and root system (described in detail in Watt et al., 2009), and Bd21-3 is the most easily transformed line (Vogel and Hill, 2008) and parent of a T-DNA mutant population (Bragg et al., 2012); c, Gaz1, Gaz4, and Gaz7; d, Kah1, Kah2, and Kah3. e, Koz1, Koz3, and Koz5; f, Tek1 and Tek6; g, exact GPS coordinates are unknown for lines Men2 (S), Mur2 (S), Bd2.3 (I), Bd3-1 (I), and Abr1 (T).  相似文献   

3.
4.
5.
6.
7.
8.
Endophytic fungi viz., Nigrospora sphaerica (E1 and E6), Subramaniula cristata (E7), and Polycephalomyces sinensis (E8 and E10) were isolated from the medicinal plant, Shirazi thyme (Zataria multiflora). In in vitro tests, these endophytes inhibited the mycelial growth of Monosporascus cannonballus, a plant pathogenic fungus. Morphological abnormalities in the hyphae of M. cannonballus at the edge of the inhibition zone in dual cultures with N. sphaerica were observed. The culture filtrates of these endophytes caused leakage of electrolytes from the mycelium of M. cannonballus. To our knowledge, this is the first report on the isolation and characterization of fungal endophytes from Z. multiflora as well as their antifungal effect on M. cannonballus.Key words: Zataria multiflora, antifungal, endophytic fungi, Monosporascus cannonballus

The term “Endophytes” denotes microorganisms that colonize plants’ internal tissues for part of or throughout their life cycle without producing any apparent adverse effect. The endophytic microorganisms include fungi, bacteria, and actinobacteria (Bacon and White 2000). Among them, fungi are the most common endophytic microorganisms (Staniek et al. 2008). Endophytic fungi are ecologically distinct polyphyletic groups of microorganisms, mostly belonging to the Ascomycota phylum (Jia et al. 2016). Several fungal endophytes have been shown to act as biological control agents for managing soil-borne plant pathogens (Toghueo et al. 2016).Zataria multiflora Boiss. (Synonyms: Zataria bracteata Boiss.; Zataria multiflora var. elatior Boiss), belonging to the Lamiaceae family is a traditional medicinal plant commonly used as a flavor ingredient in different types of foods (Sajed et al. 2013). Several medicinal properties of Z. multiflora, including antiseptic, anesthetic, antispasmodic, antioxidant, antibacterial, and immunomodulatory activities, have been documented (Sajed et al. 2013). However, studies on the endophytic microorganisms inhabiting Z. multiflora are limited (Mohammadi et al. 2016).Monosporascus cannonballus Pollack & Uecker (Ascomycota, Sordariomycetes, Diatrypaceae) is one of the most important phytopathogenic fungi causing root rot and vine decline disease in muskmelon. It causes sudden wilt and collapse of the plant at the fruiting stage, which may result in total yield loss (Martyn and Miller 1996). The fungus also infects pumpkin, cucumber, courgette, and watermelon plants (Mertely et al. 1993). The control of M. cannonballus in melon and other cucurbit crops is difficult because of the pathogen’s soil-borne nature. Earlier reports indicated that arbuscular mycorrhizal fungi (AMF) (Aleandri et al. 2015), hypovirulent isolates of M. cannonballus (Batten et al. 2000), Trichoderma spp. (Zhang et al. 1999), and antagonistic rhizobacteria (Al-Daghari et al. 2020) are effective agents for the reduction of M. cannonballus-induced root rot and vine decline of melon. In addition, it is well established that many endophytic fungi isolated from medicinal plants possess antimicrobial activity against phytopathogenic fungi (Jia et al. 2016). The objective of this study was to investigate the presence of endophytic fungi in Z. multiflora and to study theirs in vitro antagonistic activity against M. cannonballus.Z. multiflora plants (accession number 201100114) were obtained from Oman Botanic Garden, Al-Khoud, Sultanate of Oman. The plants were healthy, showing no apparent symptoms of any disease or pest infestation. A virulent isolate of M. cannonballus (ID14367), obtained from the roots of a melon plant showing root rot and vine decline (Al-Rawahi et al. 2018) was used in this study. The culture was maintained on potato dextrose agar (PDA) medium (Oxoid Ltd., Basingstoke, UK).To isolate endophytic fungi, Z. multiflora plants were washed in tap water to remove adhering soil particles. The leaves were separated, cut into small pieces, and surface-sterilized by washing in 70% (v/v) ethanol for 1 min and then in 1% (v/v) sodium hypochlorite for 1 min. The plant tissues were then washed 3–4 times with sterilized distilled water. The leaf tissue pieces were further cut into small pieces (0.2–0.5 cm in length) using a sterile scalpel and placed on PDA medium. The plates were incubated at 25 ± 2°C for 7–10 days, and pure cultures of the endophytic fungi were obtained (Lu et al. 2012).DNA was extracted from the mycelia for molecular identification of endophytic fungi according to the method described by Liu et al. (2000). PCR amplification of the Internal Transcribed Spacer (ITS) regions of the fungal rDNA was performed using the primers ITS4 (5’-TCCTCCGCTTATTGATATGC-3’) and ITS5 (5’-GGAAGTAAAAGTCGTAACAAGG-3’) as described by Halo et al. (2018). The PCR products of the expected sizes were sequenced at Macrogen, Seoul, Korea. The sequences were subjected to BLAST searches using the National Center for Biotechnology Information (NCBI) database (http://www.ncbi.nlm.nih.gov).A dual culture technique was used to test the in vitro antagonistic effect of the endophytic fungi against M. cannonballus. A mycelial plug (7-mm diameter) was excised from the fungal endophyte colonies and placed on one side of a PDA plate (90-mm diameter) about 1 cm away from the edge. On the same plate, a 7-mm diameter disc of M. cannonballus was placed on the opposite side at 1 cm distance from the edge. The Petri plates inoculated with M. cannonballus alone were used as control. Four Petri plates per treatment were used. The Petri plates were incubated at 25 ± 2°C, and the radial growth of M. cannonballus was measured after 5–7 days of incubation. The mycelial growth inhibition was calculated using the following formula: % inhibition=[1(T/C)]×100where C – radial growth of M. cannonballus in the control plate and T – radial growth of M. cannonballus in the dual culture plate (Toghueo et al. 2016).To investigate the antagonistic effects of the endophytic fungi on the morphology of M. cannonballus hyphae, the five-mm agar plug samples of M. cannonballus were excised from the colony edges of inhibition zone in the dual culture plate. The samples for scanning electron microscopy were prepared according to the method reported by Goldstein et al. (2003) and observed with a JEOL (Model: JSM-7800F) scanning electron microscope. The culture of M. cannonballus grown in the absence of endophytic fungi served as control.To perform the electrolyte leakage assay, the endophytic fungi were cultured in 200 ml of Czapek Dox broth (static) in 500 ml conical flasks at room temperature (25 ± 2°C) for 14 days, and the culture filtrates were obtained by filtering through Whatman No. 1 filter paper. Five hundred mg of M. cannonballus mycelium were added to 20 ml of culture filtrate in a glass vial. The conductivity of the suspension was measured at 0, 1, and 3 h after incubation by using a conductivity meter (Halo et al. 2018). There were three replicates per treatment and control.Data from the in vitro growth inhibition and the electrolyte leakage assays were statistically analyzed using general linear model ANOVA using Minitab Statistical Software version 17 (Minitab Inc., State College, USA). When ANOVA revealed significant differences between treatments, means were separated using Tukey’s studentized range test at p ≤ 0.05. Arc sine transformation of data on % mycelial growth inhibition was done prior to analysis.A total of five morphologically distinct fungal endophytes were obtained from the leaves of Z. multiflora. Based on the rDNA ITS sequence analysis, these endophytic fungal (Ascomycota, Sordariomycetes) isolates were identified as Nigrospora sphaerica (Amphisphaeriales, Apiosporaceae) (E1 and E6), Subramaniula cristata (Sordariales, Chaetomiaceae) (E7) and Polycephalomyces sinensis (Hypocreales, Ophiocordycipitaceae) (E8 and E10). The sequences were deposited in the GenBank database (http://www.ncbi.nlm.nih.gov/genbank/) under the accession numbers MH028052, MH028054, MH028055, MH028056, and MH028058. P. sinensis is an important medicinal fungus. Numerous pharmacological activities of P. sinensis including immunomodulatory, anti-estrogenicity and antitumor activities have been documented (Wang et al. 2012). N. sphaerica has been reported as an endophyte (Wang et al. 2017) as well as a pathogen in a few plant species (Wright et al. 2008; Liu et al. 2016). However, Z. multiflora plants colonized with these endophytic fungi were healthy and did not show any observable disease symptoms.The in vitro dual culture antagonism assay showed that all the five endophytic fungi inhibited the mycelial growth of M. cannonballus. N. sphaerica E1 was the most effective (81.7%), followed by P. sinensis E8 (80.6%), P. sinensis E10 (75.8%) and N. sphaerica E6 (66.1%). S. cristata E7 was the least effective, which recorded 38.7% inhibition (Table (TableI,I, Fig. Fig.1).1). Further, scanning electron microscopic observations of the hyphae of M. cannonballus from the dual culture assay plates at the edge of the inhibition zone revealed morphological abnormalities such as disintegration, shrinkage, and loss of turgidity. Scanning electron micrograph of M. cannonballus after co-cultivation with the endophytic fungus N. sphaerica E1 is shown in Fig. Fig.2.2. These findings corroborate with those of Hajlaoui et al. (1992) who reported plasmolysis of Sphaerotheca pannosa var. rosae mycelium due to the antagonistic effect of Sporothrix flocculosa. Halo et al. (2018) reported shrinkage of Pythium aphanidermatum hyphae due to the antagonistic activity of Aspergillus terreus. The shrinkage of M. cannonballus hyphae in the present study suggests a possible leakage of cytoplasmic contents (Garg et al. 2010). The loss of the turgidity of M. cannonballus hyphae indicates alterations in the permeability of the cell membrane (Halo et al. 2018). Several reports indicate the production of antimicrobial substances by endophytic fungi (Zhao et al. 2012; Homthong et al. 2016). Kim et al. (2001) demonstrated that phomalactone, a compound produced by N. sphaerica restricted the mycelial growth and germination of sporangium and zoospore of Phytophthora infestans and decreased the incidence of late blight in tomato. Zhao et al. (2012) characterized four secondary antifungal metabolites viz., dechlorogriseofulvin, griseofulvin, mullein, and 8-dihydroramulosin from the liquid cultures of the endophytic fungus Nigrospora sp. isolated from roots of the medicinal plant, Moringa oleifera. Homthong et al. (2016) reported the production of chitinase by Paecilomyces (Polycephalomyces) sp. The inhibitory effect of endophytic fungi on the hyphae of M. cannonballus in this study might be due to the production of antifungal metabolites.Table IPercentage inhibition of mycelial growth of M. cannonballus by endophytic fungi isolated from Zataria multiflora in dual cultures on PDA.
Fungal endophyte% Inhibition
Nigrospora sphaerica E181.7 (64.7) ± 5.5a
Nigrospora sphaerica E666.1 (54.4) ± 1.9a
Subramaniula cristata E738.7 (38.5) ± 3.7b
Polycephalomyces sinensis E880.6 (63.9) ± 11.2a
Polycephalomyces sinensis E1075.8 (60.5) ± 9.3a
Open in a separate windowData are mean of four replications ± standard deviation. Figures in parentheses are arc sine transformed values. Values in columns followed by different letters indicate significant differences according to the Tukey’s test (p < 0.05).Open in a separate windowFig. 1.In vitro growth inhibition of Monosporascus cannonballus after dual cultivation with several endophytic fungi from Zataria multiflora.a) M. cannonballus (Mc) alone; b) M. cannonballus + N. sphaerica E1; c) M. cannonballus + N. sphaerica E6; d) M. cannonballus + S. cristata E7; e) M. cannonballus + Paecilomyces sinensis E8; f) M. cannonballus + P. sinensis E10Open in a separate windowFig. 2.Scanning electron micrograph showing morphological changes in the hyphae of Monosporascus cannonballus at the edge of the inhibition zone after co-cultivation with Nigrospora sphaerica E1 in PDA platesa) Hyphae of M. cannonballus in the control; b) Hyphae of M. cannonballus after co-cultivation with N. sphaerica E1.Several reports indicate that leakage of electrolytes is an indicator of cell membrane damage in fungi (Manhas and Kaur 2016; Halo et al. 2018). The present study observed that the culture filtrates of endophytic fungi induced electrolyte leakage from the mycelium of M. cannonballus as assessed by increased conductivity of mycelial suspension upon treatment with the culture filtrates of endophytic fungi (Table (TableII).II). The maximum release of electrolytes was observed with N. sphaerica E1, followed by N. sphaerica E6, P. sinensis E10, S. cristata E7, and P. sinensis E8. The results suggest the production of antifungal metabolites as one of the possible mechanisms of action of these fungal endophytes on M. cannonballus.Table IIElectrolyte leakage induced by culture filtrates of endophytic fungi from the mycelium of M. cannonballus.
TreatmentsElectrical conductivity (mS cm–1)
0 min1 h3 h
Nigrospora sphaerica E13.95 ± 0.02a3.98 ± 0.02a4.12 ± 0.06a
Nigrospora sphaerica E63.90 ± 0.02b3.87 ± 0.02a4.01 ± 0.04b
Subramaniula cristata E73.46 ± 0.00c3.41 ± 0.01c3.55 ± 0.01d
Polycephalomyces sinensis E83.10 ± 0.03d3.28 ± 0.14c3.14 ± 0.01e
Polycephalomyces sinensis E103.50 ± 0.02c3.61 ± 0.00b3.71 ± 0.01c
Czapek Dox broth (un inoculated)2.01 ± 0.00e2.01 ± 0.00d2.08 ± 0.00f
Control (water)0.65 ± 0.01f0.67 ± 0.00e0.71 ± 0.01g
Open in a separate windowData shown correspond to mean of three replications ± the standard deviation. Values in columns followed by different letters indicate significant differences according to the Tukey’s test (p < 0.05).To our knowledge, this study is the first to report in vitro inhibitory activity of fungal endophytes isolated from Z. multiflora against M. cannonballus. Further studies are needed to evaluate the potential of these fungal endophytes in controlling root rot and vine decline disease of melon, assess their endophytic movement in melon plant, and to determine the mode of action of these fungal endophytes on M. cannonballus.  相似文献   

9.
10.
Osteoarthritis (OA) is a multidimensional health problem and a common chronic disease. It has a substantial impact on patient quality of life and is a common cause of pain and mobility issues in older adults. The functional limitations, lack of curative treatments, and cost to society all demonstrate the need for translational and clinical research. The use of OA models in mice is important for achieving a better understanding of the disease. Models with clinical relevance are needed to achieve 2 main goals: to assess the impact of the OA disease (pain and function) and to study the efficacy of potential treatments. However, few OA models include practical strategies for functional assessment of the mice. OA signs in mice incorporate complex interrelations between pain and dysfunction. The current review provides a comprehensive compilation of mouse models of OA and animal evaluations that include static and dynamic clinical assessment of the mice, merging evaluation of pain and function by using automatic and noninvasive techniques. These new techniques allow simultaneous recording of spontaneous activity from thousands of home cages and also monitor environment conditions. Technologies such as videography and computational approaches can also be used to improve pain assessment in rodents but these new tools must first be validated experimentally. An example of a new tool is the digital ventilated cage, which is an automated home-cage monitor that records spontaneous activity in the cages.

Osteoarthritis (OA) is a multidimensional health problem and a common chronic disease.36 Functional limitations, the absence of curative treatments, and the considerable cost to society result in a substantial impact on quality of life.76 Historically, OA has been described as whole joint and whole peri-articular diseases and as a systemic comorbidity.9,111 OA consists of a disruption of articular joint cartilage homeostasis leading to a catabolic pathway characterized by chondrocyte degeneration and destruction of the extracellular matrix (ECM). Low-grade chronic systemic inflammation is also actively involved in the process.42,92 In clinical practice, mechanical pain, often accompanied by a functional decline, is the main reason for consultations. Recommendations to patients provide guidance for OA management.22, 33,49,86 Evidence-based consensus has led to a variety of pharmacologic and nonpharmacologic modalities that are intended to guide health care providers in managing symptomatic patients. Animal-based research is of tremendous importance for the study of early diagnosis and treatment, which are crucial to prevent the disease progression and provide better care to patients.The purpose of animal-based OA research is 2-fold: to assess the impact of the OA disease (pain and function) and to study the efficacy of a potential treatment.18,67 OA model species include large animals such as the horse, goat, sheep, and dog, whose size and anatomy are expected to better reflect human joint conditions. However, small animals such as guinea pig, rabbit, mouse, and rat represent 77% of the species used.1,87 In recent years, mice have become the most commonly used model for studying OA. Mice have several advantageous characteristics: a short development and life span, easy and low-cost breeding and maintenance, easy handling, small joints that allow histologic analysis of the whole joint,32 and the availability of genetically modified lines.108 Standardized housing, genetically defined strains and SPF animals reduce the genetic and interindividual acquired variability. Mice are considered the best vertebrate model in terms of monitoring and controlling environmental conditions.7,14,15,87 Mouse skeletal maturation is reached at 10 wk, which theoretically constitutes the minimal age at which mice should be entered into an OA study.64,87,102 However, many studies violate this limit by testing mice at 8 wk of age.Available models for OA include the following (32,111 physical activity and exercise induced OA; noninvasive mechanical loading (repetitive mild loading and single-impact injury); and surgically induced (meniscectomy models or anterior cruciate ligament transection). The specific model used would be based on the goal of the study.7 For example, OA pathophysiology, OA progression, and OA therapies studies could use spontaneous, genetic, surgical, or noninvasive models. In addition, pain studies could use chemical models. Lastly, post-traumatic studies would use surgical or noninvasive models; the most frequently used method is currently destabilization of the medial meniscus,32 which involves transection of the medial meniscotibial ligament, thereby destabilizing the joint and causing instability-driven OA. An important caveat for mouse models is that the mouse and human knee differ in terms of joint size, joint biomechanics, and histologic characteristics (layers, cellularity),32,64 and joint differences could confound clinical translation.10 Table 1. Mouse models of osteoarthritis.
ModelsProsCons
SpontaneousWild type mice7,9,59,67,68,70,72,74,80,85,87,115,118,119,120- Model of aging phenotype
- The less invasive model
- Physiological relevance: mimics human pathogenesis
- No need for technical expertise
- No need for specific equipment
- Variability in incidence
- Large number of animals at baseline
- Long-term study: Time consuming (time of onset: 4 -15 mo)
- Expensive (husbandry)
Genetically modified mice2,7,25,40,50,52,67,72,79,80, 89,120- High incidence
- Earlier time of onset: 18 wk
- No need for specific equipment
- Combination with other models
- Time consuming for the strain development
- Expensive
Chemical- inducedMono-iodoacetate injection7,11,46,47,60,66,90,91,101,128- Model of pain-like phenotype
- To study mechanism of pain and antalgic drugs
- Short-term study: Rapid progression (2-7 wk)
- Reproducible
- Low cost
- Need for technical expertise
- Need for specific equipment
- Systemic injection is lethal
- Destructive effect: does not allow to study the early phase of pathogenesis
Papain injection66,67,120- Short-term study: rapid progression
- Low cost
- Need for technical expertise
- Need for specific equipment
- Does not mimic natural pathogenesis
Collagenase injection7,65,67,98- Short-term study: rapid progression (3 wk)
- Low cost
- Need for technical expertise
- Need for specific equipment
- Does not mimic natural pathogenesis
Non-invasiveHigh-fat diet (Alimentary induced obesity model)5,8,43,45,57,96,124Model of metabolic phenotype
No need for technical expertise
No need for specific equipment
Reproducible
Long-term study: Time consuming (8 wk–9 mo delay)
Expensive
Physical activity and exercise model45,73Model of post traumatic phenotype
No need for technical expertise
Long-term study: time consuming (18 mo delay)
Expensive
Disparity of results
Mechanical loading models Repetitive mild loading models Single-impact injury model7,16,23,24, 32,35,104,105,106Model of post traumatic phenotype
Allow to study OA development
Time of onset: 8-10 wk post injury
Noninvasive
Need for technical expertise
Need for specific equipment
Heterogeneity in protocol practices
Repetitive anesthesia required or ethical issues
SurgicalOvariectomy114Contested.
Meniscectomy model7,32,63,67,87 Model of post traumatic phenotype
High incidence
Short-term study: early time of onset (4 wk from surgery)
To study therapies
Need for technical expertise
Need for specific equipment
Surgical risks
Rapid progression compared to human
Anterior cruciate ligament transection (ACLT)7,39,40,61,48,67,70,87,126Model of posttraumatic phenotype
High incidence
Short-term study: early time of onset (3-10 wk from surgery)
Reproducible
To study therapies
Need for technical expertise
Need for specific equipment
Surgical risks
Rapid progression compared to human
Destabilization of medial meniscus (DMM)7,32,39,40Model of post traumatic phenotype
High incidence
Short-term study: early time of onset (4 wk from surgery)
To study therapies
The most frequently used method
Need for technical expertise
Need for specific equipment
Surgical risks
Rapid progression compared to human
Open in a separate windowSince all animal models have strengths and weaknesses, it is often best to plan using a number of models and techniques together to combine the results.In humans, the lack of correlation between OA imaging assessment and clinical signs highlights the need to consider the functional data and the quality of life to personalize OA management. Clinical outcomes are needed to achieve 2 main goals: to assess the impact of the OA in terms of pain and function and to study the efficacy of treatments.65 Recent reviews offer few practical approaches to mouse functional assessment and novel approaches to OA models in mice.7,32,67,75,79,83,87, 100,120 This review will focus on static and dynamic clinical assessment of OA using automatic and noninvasive emerging techniques (
Test nameTechniquesKind of assessmentOutputSpecific equipment required
Static measurement
Von Frey filament testingCalibrated nylon filaments of various thickness (and applied force) are pressed against the skin of the plantar surface of the paw in ascending order of forceStimulus- evoked pain-like behavior
Mechanical stimuli - Tactile allodynia
The most commonly used test
Latency to paw withdrawal
and
Force exerted are recorded
Yes
Knee extension testApply a knee extension on both the intact and affected knee
or
Passive extension range of the operated knee joint under anesthesia
Stimulus-evoked pain-like behaviorNumber of vocalizations evoked in 5 extensionsNone
HotplateMouse placed on hotplate. A cutoff latency has been determined to avoid lesionsStimulus-evoked pain-like behavior
Heat stimuli- thermal sensitivity
Latency of paw withdrawalYes
Righting abilityMouse placed on its backNeuromuscular screeningLatency to regain its footingNone
Cotton swab testBringing a cotton swab into contact with eyelashes, pinna, and whiskersStimulus-evoked pain-like behavior
Neuromuscular screening
Withdrawal or twitching responseNone
Spontaneous activity
Spontaneous cage activityOne by one the cages must be laid out in a specific platformSpontaneous pain behavior
Nonstimulus evoked pain
Activity
Vibrations evoked by animal movementsYes
Open field analysisExperiment is performed in a clear chamber and mice can freely exploreSpontaneous pain behavior
Nonstimulus evoked pain
Locomotor analysis
Paw print assessment
Distance traveled, average walking speed, rest time, rearing
Yes
Gait analysisMouse is placed in a specific cage equipped with a fluorescent tube and a glass plate allowing an automated quantitative gait analysisNonstimulus evoked pain
Gait analysis
Indirect nociception
Intensity of the paw contact area, velocity, stride frequency, length, symmetry, step widthYes
Dynamic weight bearing systemMouse placed is a specific cage. This method is a computerized capacitance meter (similar to gait analysis)Nonstimulus evoked pain
Weight-bearing deficits
Indirect nociception
Body weight redistribution to a portion of the paw surfaceYes
Voluntary wheel runningMouse placed is a specific cage with free access to stainless steel activity wheels. The wheel is connected to a computer that automatically record dataNonstimulus evoked pain
Activity
Distance traveled in the wheelYes
Burrowing analysisMouse placed is a specific cage equipped with steel tubes (32 cm in length and 10 cm in diameter) and quartz sand in Plexiglas cages (600 · 340x200 mm)Nonstimulus evoked pain
Activity
Amount of sand burrowedYes
Digital video recordingsMouse placed is a specific cage according to the toolNonstimulus evoked pain
Or
Evoked pain
Scale of pain or specific outcomeYes
Digital ventilated cage systemNondisrupting capacitive-based technique: records spontaneous activity 24/7, during both light and dark phases directly from the home cage rackSpontaneous pain behavior
Nonstimulus evoked pain
Activity-behavior
Distance walked, average speed, occupation front, occupation rear, activation density.
Animal locomotion index, animal tracking distance, animal tracking speed, animal running wheel distance and speed or rotation
Yes
Challenged activity
Rotarod testGradual and continued acceleration of a rotating rod onto which mice are placedMotor coordination
Indirect nociception
Rotarod latency: riding time and speed with a maximum cut off.Yes
Hind limb and fore grip strengthMouse placed over a base plate in front of a connected grasping toolMuscle strength of limbsPeak force, time resistanceYes
Wire hang analysisSuspension of the mouse on the wire and start the timeMuscle strength of limbs: muscle function and coordinationLatency to fall grippingNone
(self -constructed)
Open in a separate windowPain cannot be directly measured in rodents, so methods have been developed to quantify “pain-like” behaviors. The clinical assessment of mice should be tested both before and after the intervention (induced-OA ± administration of treatment) to take into account the habituation and establish a baseline to compare against.  相似文献   

11.
Increased Crystalline Cellulose Activity via Combinations of Amino Acid Changes in the Family 9 Catalytic Domain and Family 3c Cellulose Binding Module of Thermobifida fusca Cel9A     
Yongchao Li  Diana C. Irwin  David B. Wilson 《Applied and environmental microbiology》2010,76(8):2582-2588
Amino acid modifications of the Thermobifida fusca Cel9A-68 catalytic domain or carbohydrate binding module 3c (CBM3c) were combined to create enzymes with changed amino acids in both domains. Bacterial crystalline cellulose (BC) and swollen cellulose (SWC) assays of the expressed and purified enzymes showed that three combinations resulted in 150% and 200% increased activity, respectively, and also increased synergistic activity with other cellulases. Several other combinations resulted in drastically lowered activity, giving insight into the need for a balance between the binding in the catalytic cleft on either side of the cleavage site, as well as coordination between binding affinity for the catalytic domain and CBM3c. The same combinations of amino acid variants in the whole enzyme, Cel9A-90, did not increase BC or SWC activity but did have higher filter paper (FP) activity at 12% digestion.Cellulases catalyze the breakdown of cellulose into simple sugars that can be fermented to ethanol. The large amount of natural cellulose available is an exciting potential source of fuels and chemicals. However, the detailed molecular mechanisms of crystalline cellulose degradation by glycoside hydrolases are still not well understood and their low efficiency is a major barrier to cellulosic ethanol production.Thermobifida fusca is a filamentous soil bacterium that grows at 50°C in defined medium and can utilize cellulose as its sole carbon source. It is a major degrader of plant cell walls in heated organic materials such as compost piles and rotting hay and produces a set of enzymes that includes six different cellulases, three xylanases, a xyloglucanase, and two CBM33 binding proteins (12). Among them are three endocellulases, Cel9B, Cel6A, and Cel5A (7, 8), two exocellulases, Cel48A and Cel6B (6, 19), and a processive endocellulase, Cel9A (5, 7).T. fusca Cel9A-90 (Uniprot P26221 and YP_290232) is a multidomain enzyme consisting of a family 9 catalytic domain (CD) rigidly attached by a short linker to a family 3c cellulose binding module (CBM3c), followed by a fibronectin III-like domain and a family 2 CBM (CBM2). Cel9A-68 consists of the family 9 CD and CBM3c. The crystal structure of this species (Fig. (Fig.1)1) was determined by X-ray crystallography at 1.9 Å resolution (Protein Data Bank [PDB] code 4tf4) (15). Previous work has shown that E424 is the catalytic acid and D58 is the catalytic base (11, 20). H125 and Y206 were shown to play an important role in activity by forming a hydrogen bonding network with D58, an important supporting residue, D55, and Glc(−1)O1. Several enzymes with amino acid changes in subsites Glc(−1) to Glc(−4) had less than 20% activity on bacterial cellulose (BC) and markedly reduced processivity. It was proposed that these modifications disturb the coordination between product release and the subsequent binding of a cellulose chain into subsites Glc(−1) to Glc(−4) (11). Another variant enzyme with a deletion of a group of amino acids forming a block at the end of the catalytic cleft, Cel9A-68 Δ(T245-L251)R252K (DEL), showed slightly improved filter paper (FP) activity and binding to BC (20).Open in a separate windowFIG. 1.Crystal structure of Cel9A-68 (PDB code 4tf4) showing the locations of the variant residues, catalytic acid E424, catalytic base D58, hydrogen bonding network residues D55, H125, and Y206, and six glucose residues, Glc(−4) to Glc(+2). Part of the linker is visible in dark blue.The CBM3c domain is critical for hydrolysis and processivity. Cel9A-51, an enzyme with the family 9 CD and the linker but without CBM3c, had low activity on carboxymethyl cellulose (CMC), BC, and swollen cellulose (SWC) and showed no processivity (4). The role of CBM3c was investigated by mutagenesis, and one modified enzyme, R557A/E559A, had impaired activity on all of these substrates but normal binding and processivity (11). Variants with changes at five other CBM3c residues were found to slightly lower the activity of the modified enzymes, while Cel9A-68 enzymes containing either F476A, D513A, or I514H were found to have slightly increased binding and processivity (11) (see Table Table1).1). In the present work, CBM3c has been investigated more extensively to identify residues involved in substrate binding and processivity, understand the role of CBM3c more clearly, and study the coordination between the CD and CBM3c. An additional goal was to combine amino acid variants showing increased crystalline cellulose activity to see if this further increased activity. Finally, we have investigated whether the changes that improved the activity of Cel9A-68 also enhanced the activity of intact Cel9A-90.

TABLE 1.

Activities of Cel9A-68 CBM3c variant enzymes and CD variant enzymes used to create the double variants
EnzymeActivity (% of wild type) on:
% Processivity% BC bindingReference
CMCSWCBCFPa
Wild type10010010010010015This work
R378K9891103931392011
DELb981011011289620
F476A97105791001452111
D513A1001151211071192011
I514H104911121041102311
Y520A1087833a79871411
R557A1039860a9390This work
E559A869030a7094This work
R557A+E559A907515a751061511
Q561A1035651a7874This work
R563A977052a931292011
Open in a separate windowaThe target percent digestion could not be reached; activity was calculated using 1.5 μM enzyme.bDEL refers to deletion of T245 to L251 and R252K.  相似文献   

12.
Stress-induced Ceramide-activated Protein Phosphatase Can Compensate for Loss of Amphiphysin-like Activity In Saccharomyces cerevisiae and Functions to Reinitiate Endocytosis     
Paula C. McCourt  Jeanelle M. Morgan    Joseph T. Nickels  Jr. 《The Journal of biological chemistry》2009,284(18):11930-11941
  相似文献   

13.
Loss of Par-1a/MARK3/C-TAK1 Kinase Leads to Reduced Adiposity,Resistance to Hepatic Steatosis,and Defective Gluconeogenesis     
Jochen K. Lennerz  Jonathan B. Hurov  Lynn S. White  Katherine T. Lewandowski  Julie L. Prior  G. James Planer  Robert W. Gereau  IV  David Piwnica-Worms  Robert E. Schmidt  Helen Piwnica-Worms 《Molecular and cellular biology》2010,30(21):5043-5056
Par-1 is an evolutionarily conserved protein kinase required for polarity in worms, flies, frogs, and mammals. The mammalian Par-1 family consists of four members. Knockout studies of mice implicate Par-1b/MARK2/EMK in regulating fertility, immune homeostasis, learning, and memory as well as adiposity, insulin hypersensitivity, and glucose metabolism. Here, we report phenotypes of mice null for a second family member (Par-1a/MARK3/C-TAK1) that exhibit increased energy expenditure, reduced adiposity with unaltered glucose handling, and normal insulin sensitivity. Knockout mice were protected against high-fat diet-induced obesity and displayed attenuated weight gain, complete resistance to hepatic steatosis, and improved glucose handling with decreased insulin secretion. Overnight starvation led to complete hepatic glycogen depletion, associated hypoketotic hypoglycemia, increased hepatocellular autophagy, and increased glycogen synthase levels in Par-1a−/− but not in control or Par-1b−/− mice. The intercrossing of Par-1a−/− with Par-1b−/− mice revealed that at least one of the four alleles is necessary for embryonic survival. The severity of phenotypes followed a rank order, whereby the loss of one Par-1b allele in Par-1a−/− mice conveyed milder phenotypes than the loss of one Par-1a allele in Par-1b−/− mice. Thus, although Par-1a and Par-1b can compensate for one another during embryogenesis, their individual disruption gives rise to distinct metabolic phenotypes in adult mice.Cellular polarity is a fundamental principle in biology (6, 36, 62). The prototypical protein kinase originally identified as a regulator of polarity was termed partitioning defective (Par-1) due to early embryonic defects in Caenorhabditis elegans (52). Subsequent studies revealed that Par-1 is required for cellular polarity in worms, flies, frogs, and mammals (4, 17, 58, 63, 65, 71, 89). An integral role for Par-1 kinases in multiple signaling pathways has also been established, and although not formally addressed, multifunctionality for individual Par-1 family members is implied in reviews of the list of recognized upstream regulators and downstream substrates (Table (Table1).1). Interestingly, for many Par-1 substrates the phosphorylated residues generate 14-3-3 binding sites (25, 28, 37, 50, 59, 61, 68, 69, 78, 95, 101, 103). 14-3-3 binding in turn modulates both nuclear/cytoplasmic as well as cytoplasmic/membrane shuttling of target proteins, thus allowing Par-1 activity to establish intracellular spatial organization (15, 101). The phosphorylation of Par-1 itself promotes 14-3-3 binding, thereby regulating its subcellular localization (37, 59, 101).

TABLE 1.

Multifunctionality of Par-1 polarity kinase pathwaysa
Regulator or substrateFunctionReference(s)
Regulators (upstream function)
    LKB1Wnt signaling, Peutz-Jeghers syndrome, insulin signal transduction, pattern formation2, 63, 93
    TAO1MEK3/p38 stress-responsive mitogen-activated protein kinase (MAPK) pathway46
    MARKKNerve growth factor signaling in neurite development and differentiation98
    aPKCCa2+/DAG-independent signal transduction, cell polarity, glucose metabolism14, 37, 40, 45, 59, 75, 95
    nPKC/PKDDAG-dependent, Ca2+-independent signal transduction (GPCR)101
    PAR-3/PAR-6/aPKC(−); regulates Par-1, assembly of microtubules, axon-dendrite specification19
    GSK3β(−); tau phosphorylation, Alzheimer''s dementia, energy metabolism, body patterning54, 97
    Pim-1 oncogene(−); G2/M checkpoint, effector of cytokine signaling and Jak/STAT(3/5)5
    CaMKI(−); Ca2+-dependent signal transduction, neuronal differentiation99
Substrates (downstream function)
    Cdc25CRegulation of mitotic entry by activation of the cdc2-cyclin B complex25, 72, 78, 103
    Class II HDACControl of gene expression and master regulator of subcellular trafficking28, 50
    CRTC2/TORC2Gluconeogenesis regulator via LKB1/AMPK/TORC2 signaling, PPARγ1a coactivator49
    Dlg/PSD-95Synaptogenesis and neuromuscular junction, tumor suppressor (102)104
    DisheveledWnt signaling, translocation of Dsh from cytoplasmic vesicles to cortex73, 94
    KSR1Regulation of the Ras-MAPK pathway68, 69
    MAP2/4/TAUDynamic instability (67, 83) of microtubules, Alzheimer''s dementia (30)11, 31-33, 47, 70, 96
    Mib/NotchMind bomb (Mib degradation and repression of Notch signaling results in neurogenesis)57, 74, 81
    Par3/OSKAR/LglCytoplasmic protein segregation, cell polarity, and asymmetric cell division7, 10
    Pkp2Desmosome assembly and organization; nuclear shuttling68, 69
    PTPH1Linkage between Ser/Thr and Tyr phosphorylation-dependent signaling103
    Rab11-FIPRegulation of endocytosis (23), trafficking of E-cadherin (64)34
Open in a separate windowaLKB1 also is known as Par-4; MARKK also is known as Ste20-like; (−), inhibitory/negative regulation has been shown; GPCR, G protein-coupled receptors. MARKK is highly homologous to TAO-1 (thousand-and-one amino acid kinase) (46).The mammalian Par-1 family contains four members (Table (Table2).2). Physiological functions of the Par-1b kinase have been studied using targeted gene knockout approaches in mice (9, 44). Two independently derived mouse lines null for Par-1b have implicated this protein kinase in diverse physiological processes, including fertility (9), immune system homeostasis (44), learning and memory (86), the positioning of nuclei in pancreatic beta cells (35, 38), and growth and metabolism (43).

TABLE 2.

Terminology and localization of mammalian Par-1 family members
SynonymsaSubcellular localization
Par-1a, MARK3, C-TAK1, p78/KP78, 1600015G02Rik, A430080F22Rik, Emk2, ETK-1, KIAA4230, mKIAA1860, mKIAA4230, M80359Basolateralb/apicalc
Par-1b, EMK, MARK2, AU024026, mKIAA4207Basolateral
Par1c, MARK1Basolateral
Par1d, MARK4, MARKL1Not asymmetricd
Open in a separate windowaPar should not to be confused with protease-activated receptor 1 (PAR1 [29]); C-TAK1, Cdc twenty-five C-associated kinase 1; MARK, microtubule affinity regulating kinase; MARKL, MAP/microtubule affinity-regulating kinase-like 1.bBasolateral to a lesser degree than Par-1b (37).cHuman KP78 is asymmetrically localized to the apical surface of epithelial cells (76).dVariant that does not show asymmetric localization in epithelial cells when overexpressed (95).Beyond Par-1b, most information regarding the cell biological functions of the Par-1 kinases comes from studies of Par-1a. Specifically, Par-1a has been implicated in pancreatic (76) and hepatocarcinogenesis (51), as well as colorectal tumors (77), hippocampal function (100), CagA (Helicobacter pylori)-associated epithelial cell polarity disruption (82), and Peutz-Jeghers syndrome (48), although the latter association has been excluded recently (27). As a first step toward determining unique and redundant functions of Par-1 family members, mice disrupted for a second member of the family (Par-1a/MARK3/C-TAK1) were generated. We report that Par-1a−/− mice are viable and develop normally, and adult mice are hypermetabolic, have decreased white and brown adipose tissue mass, and unaltered glucose/insulin handling. However, when challenged by a high-fat diet (HFD), Par-1a−/− mice exhibit resistance to hepatic steatosis, resistance to glucose intolerance, and the delayed onset of obesity relative to that of control littermates. Strikingly, overnight starvation results in a complete depletion of glycogen and lipid stores along with an increase in autophagic vacuoles in the liver of Par-1a−/− but not Par-1b−/− mice. Correspondingly, Par-1a−/− mice develop hypoketotic hypoglycemia. These findings reveal unique metabolic functions of two Par-1 family members.  相似文献   

14.
Evidence for Multiple Recent Host Species Shifts among the Ranaviruses (Family Iridoviridae)     
James K. Jancovich  Michel Bremont  Jeffrey W. Touchman  Bertram L. Jacobs 《Journal of virology》2010,84(6):2636-2647
  相似文献   

15.
Natural Infection of Burkholderia pseudomallei in an Imported Pigtail Macaque (Macaca nemestrina) and Management of the Exposed Colony     
Crystal H Johnson  Brianna L Skinner  Sharon M Dietz  David Blaney  Robyn M Engel  George W Lathrop  Alex R Hoffmaster  Jay E Gee  Mindy G Elrod  Nathaniel Powell  Henry Walke 《Comparative medicine》2013,63(6):528-535
Identification of the select agent Burkholderia pseudomallei in macaques imported into the United States is rare. A purpose-bred, 4.5-y-old pigtail macaque (Macaca nemestrina) imported from Southeast Asia was received from a commercial vendor at our facility in March 2012. After the initial acclimation period of 5 to 7 d, physical examination of the macaque revealed a subcutaneous abscess that surrounded the right stifle joint. The wound was treated and resolved over 3 mo. In August 2012, 2 mo after the stifle joint wound resolved, the macaque exhibited neurologic clinical signs. Postmortem microbiologic analysis revealed that the macaque was infected with B. pseudomallei. This case report describes the clinical evaluation of a B. pseudomallei-infected macaque, management and care of the potentially exposed colony of animals, and protocols established for the animal care staff that worked with the infected macaque and potentially exposed colony. This article also provides relevant information on addressing matters related to regulatory issues and risk management of potentially exposed animals and animal care staff.Abbreviations: CDC, Centers for Disease Control and Prevention; IHA, indirect hemagglutination assay; PEP, postexposure prophylacticBurkholderia pseudomallei, formerly known as Pseudomonas pseudomallei, is a gram-negative, aerobic, bipolar, motile, rod-shaped bacterium. B. pseudomallei infections (melioidosis) can be severe and even fatal in both humans and animals. This environmental saprophyte is endemic to Southeast Asia and northern Australia, but it has also been found in other tropical and subtropical areas of the world.7,22,32,42 The bacterium is usually found in soil and water in endemic areas and is transmitted to humans and animals primarily through percutaneous inoculation, ingestion, or inhalation of a contaminated source.8, 22,28,32,42 Human-to-human, animal-to-animal, and animal-to-human spread are rare.8,32 In December 2012, the National Select Agent Registry designated B. pseudomallei as a Tier 1 overlap select agent.39 Organisms classified as Tier 1 agents present the highest risk of deliberate misuse, with the most significant potential for mass casualties or devastating effects to the economy, critical infrastructure, or public confidence. Select agents with this status have the potential to pose a severe threat to human and animal health or safety or the ability to be used as a biologic weapon.39Melioidosis in humans can be challenging to diagnose and treat because the organism can remain latent for years and is resistant to many antibiotics.12,37,41 B. pseudomallei can survive in phagocytic cells, a phenomenon that may be associated with latent infections.19,38 The incubation period in naturally infected animals ranges from 1 d to many years, but symptoms typically appear 2 to 4 wk after exposure.13,17,35,38 Disease generally presents in 1 of 2 forms: localized infection or septicemia.22 Multiple methods are used to diagnose melioidosis, including immunofluorescence, serology, and PCR analysis, but isolation of the bacteria from blood, urine, sputum, throat swabs, abscesses, skin, or tissue lesions remains the ‘gold standard.’9,22,40,42 The prognosis varies based on presentation, time to diagnosis, initiation of appropriate antimicrobial treatment, and underlying comorbidities.7,28,42 Currently, there is no licensed vaccine to prevent melioidosis.There are several published reports of naturally occurring melioidosis in a variety of nonhuman primates (NHP; 2,10,13,17,25,30,31,35 The first reported case of melioidosis in monkeys was recorded in 1932, and the first published case in a macaque species was in 1966.30 In the United States, there have only been 7 documented cases of NHP with B. pseudomallei infection.2,13,17 All of these cases occurred prior to the classification of B. pseudomallei as a select agent. Clinical signs in NHP range from subclinical or subacute illness to acute septicemia, localized infection, and chronic infection. NHP with melioidosis can be asymptomatic or exhibit clinical signs such as anorexia, wasting, purulent drainage, subcutaneous abscesses, and other soft tissue lesions. Lymphadenitis, lameness, osteomyelitis, paralysis and other CNS signs have also been reported.2,7,10,22,28,32 In comparison, human''s clinical signs range from abscesses, skin ulceration, fever, headache, joint pain, and muscle tenderness to abdominal pain, anorexia, respiratory distress, seizures, and septicemia.7,9,21,22

Table 1.

Summary of reported cases of naturally occurring Burkholderia pseudomalleiinfections in nonhuman primates
CountryaImported fromDate reportedSpeciesReference
AustraliaBorneo1963Pongo sp.36
BruneiUnknown1982Orangutan (Pongo pygmaeus)33
France1976Hamlyn monkey (Cercopithecus hamlyni) Patas monkey (Erythrocebus patas)11
Great BritainPhilippines and Indonesia1992Cynomolgus monkey (Macaca fascicularis)10
38
MalaysiaUnknown1966Macaca spp.30
Unknown1968Spider monkey (Brachytelis arachnoides) Lar gibbon (Hylobates lar)20
Unknown1969Pig-tailed macaque (Macaca nemestrina)35
Unknown1984Banded leaf monkey (Presbytis melalophos)25
SingaporeUnknown1995Gorillas, gibbon, mandrill, chimpanzee43
ThailandUnknown2012Monkey19
United StatesThailand1970Stump-tailed macaque (Macaca arctoides)17
IndiaPig-tailed macaque (Macaca nemestrina)
AfricaRhesus macaque (Macaca mulatta) Chimpanzee (Pan troglodytes)
Unknown1971Chimpanzee (Pan troglodytes)3
Malaysia1981Pig-tailed macaque (Macaca nemestrina)2
Wild-caught, unknown1986Rhesus macaque (Macaca mulatta)13
Indonesia2013Pig-tailed macaque (Macaca nemestrina)Current article
Open in a separate windowaCountry reflects the location where the animal was housed at the time of diagosis.Here we describe a case of melioidosis diagnosed in a pigtail macaque (Macaca nemestrina) imported into the United States from Indonesia and the implications of the detection of a select agent identified in a laboratory research colony. We also discuss the management and care of the exposed colony, zoonotic concerns regarding the animal care staff that worked with the shipment of macaques, effects on research studies, and the procedures involved in reporting a select agent incident.  相似文献   

16.
Evidence for Interspecies Gene Transfer in the Evolution of 2,4-Dichlorophenoxyacetic Acid Degraders     
Catherine McGowan  Roberta Fulthorpe  Alice Wright  J. M. Tiedje 《Applied and environmental microbiology》1998,64(10):4089-4092
Small-subunit ribosomal DNA (SSU rDNA) from 20 phenotypically distinct strains of 2,4-dichlorophenoxyacetic acid (2,4-D)-degrading bacteria was partially sequenced, yielding 18 unique strains belonging to members of the alpha, beta, and gamma subgroups of the class Proteobacteria. To understand the origin of 2,4-D degradation in this diverse collection, the first gene in the 2,4-D pathway, tfdA, was sequenced. The sequences fell into three unique classes found in various members of the beta and gamma subgroups of Proteobacteria. None of the α-Proteobacteria yielded tfdA PCR products. A comparison of the dendrogram of the tfdA genes with that of the SSU rDNA genes demonstrated incongruency in phylogenies, and hence 2,4-D degradation must have originated from gene transfer between species. Only those strains with tfdA sequences highly similar to the tfdA sequence of strain JMP134 (tfdA class I) transferred all the 2,4-D genes and conferred the 2,4-D degradation phenotype to a Burkholderia cepacia recipient.Bacteria capable of mineralizing 2,4-dichlorophenoxyacetic acid (2,4-D), a commonly used herbicide, are found in many different phylogenetic groups (2, 3, 7, 11, 22, 23). Evidence suggests that numerous variants of 2,4-D catabolic genes exist and that catabolic operons consist of a near-random mixing of these variants (7). Interspecies gene transfer is a well-documented phenomenon (13), and horizontal gene transfer of the 2,4-D-degrading plasmid pJP4 has been shown (3, 5). However, not all 2,4-D catabolic operons are found on plasmids (10, 11, 16, 20). The extent to which other 2,4-D genes have been exchanged in nature is unknown. The aim of this research was to assess the role of horizontal gene transfer in the evolution of 2,4-D-degrading strains. This article summarizes the results of two aspects of this work—the study of the transfer of the entire 2,4-D pathway by using standard mating experiments and a phylogenetic study of the tfdA gene. The tfdA gene codes for an α-ketoglutarate-dependent 2,4-D dioxygenase which converts 2,4-D into 2,4-dichlorophenol and glyoxylate (6). This 861-bp gene was first sequenced from Ralstonia eutropha JMP134 (19). Two more tfdA genes were cloned from chromosomal locations in Burkholderia strain RASC and Burkholderia strain TFD6 (16, 20). These proved to be identical to each other and 78.5% similar to the original. An alignment of the two variants allowed conserved areas to be identified and primers to be designed for the amplification of tfdA-like genes from other sources (24). Sequence analysis of putative tfdA fragments and the small-subunit ribosomal DNA (SSU rDNA) of the strains carrying them allowed us to construct phylogenies of the genes and their hosts and to look for congruency between them.

Mating experiments.

A collection of 2,4-D degraders containing 15 unique strains as determined by genomic fingerprinting (7) was used as a source of donors in a series of mating experiments (Table (Table1).1). Burkholderia cepacia D5, lacking the ability to grow on 2,4-D and not hybridizing to any tfd genes, was used as a recipient in mating experiments. Strain D5 contains neomycin phosphotransferase genes (nptII) carried on transposon Tn5 and is resistant to 50 μg each of kanamycin, carbenicillin, and bacitracin per ml. All of the 2,4-D strains used were sensitive to these antibiotics. Filter matings were performed with a donor-to-recipient ratio of 1:10. Colonies which grew on selective medium (500 ppm of 2,4-D in mineral salts agar [MMO] [23] including 50 μg of kanamycin, carbenicillin, and bacitracin per ml) were subjected to further tests. Their ability to catabolize 2,4-D was tested in liquid medium (same composition as that described above).

TABLE 1

2,4-D-degrading strains, geographic origins, and GenBank accession numbers
StrainGenBank accession no. (SSU rDNA)OriginMost similar to genus and/or speciesaTransferbtfdA typecGenBank accession no. (tfdA gene)Reference or source
JMP134AF049542AustraliaRalstonia eutropha+IM167303
EML1549AF049546OregonBurkholderia sp.+I2
TFD39AF049539SaskatchewanBurkholderia sp.+IU4319723
K712AF049543MichiganBurkholderia sp.+IU4327611
TFD9AF049537SaskatchewanAlcaligenes xylosoxidans+IU4327623
TFD41AF049541MichiganRalstonia eutropha+I23
TFD38AF049540MichiganRalstonia eutropha+NDc23
TFD23AF049536MichiganRhodoferax fermentans+IU4327623
RASCAF049544OregonBurkholderia sp.(+)IIU257172
TFD6AF049546MichiganBurkholderia sp.II23
TFD2AF049545MichiganBurkholderia sp.II23
TFD31AF049536SaskatchewanRhodoferax fermentansIII23
B6-9AF049538OntarioRhodoferax fermentansNDIIIU431969
I-18U22836OregonHalomonas sp.NDIIIU2249915
K1443AF049531MichiganSphingomonas sp.d11
2,4-D1AF049535MontanaSphingomonas sp.R. Sanford
B6-5AF049533OntarioSphingomonas sp.ND9
B6-10AF049534OntarioSphingomonas sp.ND9
EML146AF049532OregonSphingomonas sp.2
M1AF049530French PolynesiaRhodospeudomonas sp.NDR. Fulthorpe
Open in a separate windowaThe generus and/or species most similar to the strain is given based on similarities of SSU rDNA sequences. bSymbols: +, able to transfer 2,4-D degradation to B. cepacia D5; (+), able to transfer at very low frequency; −, no transfer detected. cND, not determined. d—, no amplificate was obtained. The disappearance of 2,4-D from the culture medium was monitored by high-performance liquid chromatography. Cells were removed by centrifugation, and the supernatant was filtered through 0.2-μm-pore-size filters. These samples were then analyzed on a Lichrosorb Rp-18 column (Anspec Co., Ann Arbor, Mich.) with 60% methanol–40% 0.1% H3PO4 as the eluant. 2,4-D was detected by measuring light absorption at 230 nm. The presence of tfd genes was detected by hybridizing colony blots with a DNA probe derived from the entire pJP4 plasmid. The identity of the colonies was confirmed by probing with the nptII gene of Tn5 (found in B. cepacia D5). Probes were labeled with random hexanucleotides incorporating [32P]dCTP (3,000 Ci/mmol; New England Nuclear, Boston, Mass.). Hybridizations were done under high-stringency conditions by using 50% formamide and Denhardt’s solution (18) at 42°C. Of the 15 unique strains tested, 9 transferred 2,4-D degradation abilities to D5. This transfer was confirmed by hybridization with pJP4 for eight of these strains. B. cepacia RASC could transfer degradative abilities, but neither it nor the transconjugant hybridized to the pJP4 probe. Work subsequent to this study has confirmed that the genes carried by RASC do not hybridize to those found on pJP4 under high-stringency conditions (7).

Phylogenetic analyses.

Total genomic DNA was isolated from 20 unique 2,4-D-degrading strains (including all 15 used for mating experiments) grown on 500 ppm of 2,4-D mineral salts medium amended with 50 ppm of yeast extract. SSU rDNA was amplified by using fD1 and rD1 as primers (25). Putative tfdA fragments were amplified by using primers TVU and TVL as previously described (24). PCR products were purified with a Gene Clean kit (Bio 101, La Jolla, Calif.). Sequencing was done with an Applied Biosystems model 373A automatic sequencer (Perkin-Elmer Cetus) by using fluorescently labeled dye termination at the Michigan State University Sequencing Facility. The sequencing primer used for SSU rDNA fragments was 519R (5′ GTA TTA CCG CGG CTG CTG G-3′). For tfdA fragments, the sequencing primers were the same as the amplification primers. GenBank accession numbers for these sequences are given in Table Table11.The SSU rDNA sequences were compared to sequences in GenBank by using the Basic Local Alignment Search Tool (BLAST) (1), and those strains with the highest maximal segment pair scores were retrieved from GenBank and included in the phylogenetic analysis. Sequences were aligned manually with the software SeqEd (Applied Biosystems) and with MacClade (14). Sites where nucleotides were not resolved for all sequences were deleted from the alignment, as were those nucleotides corresponding to the small loop in this region that is absent in the alpha subgroup of the class Proteobacteria. These deletions left 283 unambiguous sites for the construction of the SSU rDNA phylogenies. Phylogenetic trees were constructed by using the neighbor-joining analysis of pairwise Jukes-Cantor distances (4), and the topology was confirmed by using the maximum parsimony method PAUP (21). Desulfomonile tiedjei of the δ-Proteobacteria was used as an outgroup. Bootstrap analysis based on 100 replicates was used to place confidence estimates on the tree. Only bootstrap values of greater than 50 were used.

2,4-D degrader diversity.

The 2,4-D degraders in this study were distributed throughout the alpha, beta, and gamma subgroups of the Proteobacteria (Fig. (Fig.1).1). The lack of representation of gram-positive bacteria is likely a reflection of isolation methods, not of the lack of gram-positive 2,4-D degraders. The majority of these strains were members of the beta subgroup of Proteobacteria, five of which were most closely related to the genus Burkholderia, having at least 92% sequence similarity with each other. Three were closely related to Rhodoferax fermentans (close to the class Comamonadaceae), three were related to Ralstonia eutropha, and one was related to Alcaligenes xylosoxidans. TFD39 falls outside any clear cluster. One member of the γ-Proteobacteria, strain I-18, a haloalkaliphile, was found to be closely related to the salt-loving genus Halomonas (15). The remaining six strains all clustered in the alpha branch of Proteobacteria (Fig. (Fig.1).1). Of this subgroup, five were most closely related to the genus Sphingomonas. One member of the α-Proteobacteria, strain M1, which is the most oligotrophic and slow growing of all the strains used in this study, is 97% similar to Rhodopseudomonas palustris. The character of strain M1 correlates well with its phylogenetic placement near the slow-growing genus Bradyrhizobium. Open in a separate windowFIG. 1Neighbor-joining dendrogram (Jukes-Cantor distances) of SSU rDNA from 2,4-D-degrading bacteria (indicated in boldface type) and reference strains (indicated in italic type). Class I (•), class II (▴), and class III (■) types of tfdA genes are indicated. Bootstrap confidence limits (percentages) are indicated above each branch. Scale bar represents a Jukes-Cantor distance of 0.01.

tfdA gene fragments.

tfdA gene fragments were successfully amplified and sequenced from 10 strains of β-Proteobacteria and 1 strain of γ-Protobacteria. None of the strains from the α-Proteobacteria gave any amplificates with these primers. These 313 contiguous nucleotides were aligned with additional tfdA sequences from JMP134 and from strain RASC (Fig. (Fig.2).2). Three distinct classes of tfdA gene sequences with slight variations in each class were found. Class I included fragments from JMP134, TFD39, TFD23, K712, and TFD9 that differed from each other by 2 bp at the most. Class I tfdA genes are probably plasmid encoded. All strains with a class I tfdA gene examined so far contained broad-host-range, self-transmissible plasmids containing 2,4-D genes (2, 3, 11, 17). All of the strains with a class I tfdA gene were able to transfer the 2,4-D phenotype in the mating studies reported above. The class II tfdA sequences included identical fragments amplified from RASC, TFD6, and TFD2 which were 76% similar to those in class I. Class III included identical fragments from strains TFD31, B6-9, and I-18 which were 77% similar to class I genes and 80% similar to class II genes. Both class II and III tfdA genes differed from each other and from class I genes in the same nine sites corresponding to the third base pair of the codons. The tfdA phylogenetic tree is a simple one, with three distinct branches that are incongruent with the SSU rDNA-derived phylogeny (Fig. (Fig.3).3). Class I tfdA sequences were found in Burkholderia-like strains, in strains related to the Comamonas-Rhodoferax group, and in the Ralstonia-Acaligenes group, all in the β-Proteobacteria. Class II sequences are less widely distributed, found only in Burkholderia-like branches. However, even in this subgroup, this tfdA variant is found in strains that differ by 7% at the SSU rDNA level (RASC and TFD2). However, the class III sequences were most interesting, being found both in the Comamonas-Rhodoferax group and in a strain of the γ-Proteobacteria, I-18, strains that differ by 24% at the SSU rDNA level. Class III genes have since been found in a collection of randomly isolated non-2,4-D degraders, including gram-positive bacilli, as well as in various gram-negative bacteria, even though the gene is not expressed (10). Open in a separate windowFIG. 2Alignment of 313 nucleotides of internal fragments of tfdA genes from representative strains. Nucleotides identical to tfdA from pJP4 are represented by periods.Open in a separate windowFIG. 3Phylogenetic incongruency of tfdA genes and SSU rDNA from diverse 2,4-D-degrading bacteria. Dendrograms for tfdA and SSU rDNA are indicated. Shading indicates the type of tfdA sequence, either class I, II, or III. Note that branch lengths are not drawn to scale.An interesting result was the detection of two different tfdA gene variants in sibling strains. TFD23 and TFD31 are identical at the ribosomal gene level, but one harbors a class I gene and the other harbors a class III gene. Similarly, TFD6 and EML159 are rRNA siblings that carry a class II and class I gene, respectively.None of the α-Proteobacteria yielded a PCR product when amplified with the conserved tfdA primers. This finding complements our observation that none of these bacteria hybridized to the tfdA gene, even under conditions of low stringency, indicating that any tfdA-like genes in the α-Proteobacteria are likely to be more divergent from the ones sequenced here (7, 11). In addition, none of the Sphingomonas strains in the study hybridized with a whole pJP4 probe, and similarly, no Sphingomonas strains scored positive for transfer of 2,4-D-degrading ability to recipient B. cepacia D5. Together these results suggest a reduced gene flow between members of the α- and β- or γ-Proteobacteria or poor gene expression of β- or γ-derived genes by α-Proteobacteria. Although plasmid pJP4 is a broad-host-range plasmid and has been known to transfer to α-Proteobacteria such as Rhizobium and Agrobacterium species and to γ-Proteobacteria such as Pseudomonas putida, Pseudomonas fluorescens, and Pseudomonas aeruginosa, the 2,4-D pathway is not expressed in these strains of the α- or γ-Proteobacteria (3). Phylogenetically limited expression of plasmid-borne 3-chlorobenzoate-degradative genes has also been noted for the pseudomonads (8). Subsequent studies have found divergent but related sequences for the tfdB and tfdC genes in 2,4-D-degrading Sphingomonas strains (7, 12, 24).With the exceptions of the minor differences within the class I pJP4-like tfdA sequences, there were no intermediate tfdA sequences. The most likely explanation of this is that the rate of horizontal transfer of the tfd genes is high relative to the rate at which mutations can accumulate. Examination of sequences of tfdA genes from a greater variety of organisms may turn up more intermediate variation.  相似文献   

17.
Open Labware: 3-D Printing Your Own Lab Equipment     
Tom Baden  Andre Maia Chagas  Greg Gage  Timothy Marzullo  Lucia L. Prieto-Godino  Thomas Euler 《PLoS biology》2015,13(3)
The introduction of affordable, consumer-oriented 3-D printers is a milestone in the current “maker movement,” which has been heralded as the next industrial revolution. Combined with free and open sharing of detailed design blueprints and accessible development tools, rapid prototypes of complex products can now be assembled in one’s own garage—a game-changer reminiscent of the early days of personal computing. At the same time, 3-D printing has also allowed the scientific and engineering community to build the “little things” that help a lab get up and running much faster and easier than ever before.Applications of 3-D printing technologies (Fig. 1A, Box 1) have become as diverse as the types of materials that can be used for printing. Replacement parts at the International Space Station may be printed in orbit from durable plastics or metals, while back on Earth the food industry is starting to explore the same basic technology to fold strings of chocolate into custom-shaped confectionary. Also, consumer-oriented laser-cutting technology makes it very easy to cut raw materials such as sheets of plywood, acrylic, or aluminum into complex shapes within seconds. The range of possibilities comes to light when those mechanical parts are combined with off-the-shelf electronics, low-cost microcontrollers like Arduino boards [1], and single-board computers such as a Beagleboard [2] or a Raspberry Pi [3]. After an initial investment of typically less than a thousand dollars (e.g., to set-up a 3-D printer), the only other materials needed to build virtually anything include a few hundred grams of plastic (approximately US$30/kg), cables, and basic electronic components [4,5].Open in a separate windowFig 1Examples of open 3-D printed laboratory tools. A 1, Components for laboratory tools, such as the base for a micromanipulator [18] shown here, can be rapidly prototyped using 3-D printing. A 2, The printed parts can be easily combined with an off-the-shelf continuous rotation servo-motor (bottom) to motorize the main axis. B 1, A 3-D printable micropipette [8], designed in OpenSCAD [19], shown in full (left) and cross-section (right). B 2, The pipette consists of the printed parts (blue), two biro fillings with the spring, an off-the-shelf piece of tubing to fit the tip, and one screw used as a spacer. B 3, Assembly is complete with a laboratory glove or balloon spanned between the two main printed parts and sealed with tape to create an airtight bottom chamber continuous with the pipette tip. Accuracy is ±2–10 μl depending on printer precision, and total capacity of the system is easily adjusted using two variables listed in the source code, or accessed via the “Customizer” plugin on the thingiverse link [8]. See also the first table.

Box 1. Glossary

Open source

A collective license that defines terms of free availability and redistribution of published source material. Terms include free and unrestricted distribution, as well as full access to source code/blueprints/circuit board designs and derived works. For details, see http://opensource.org.

Maker movement

Technology-oriented extension of the traditional “Do-it-Yourself (DIY)” movement, typically denoting specific pursuits in electronics, CNC (computer numerical control) tools such as mills and laser cutters, as well as 3-D printing and related technologies.

3-D printing

Technology to generate three-dimensional objects from raw materials based on computer models. Most consumer-oriented 3-D printers print in plastic by locally melting a strand of raw material at the tip (“hot-end”) and “drawing” a 3-D object in layers. Plastic materials include Acrylnitrile butadiene styrene (ABS) and Polylactic acid (PLA). Many variations of 3-D printers exist, including those based on laser-polymerization or fusion of resins or powdered raw materials (e.g., metal or ceramic printers).

Arduino boards

Inexpensive and consumer-oriented microcontroller boards built around simple processors. These boards offer a variety of interfaces (serial ports, I2C and CAN bus, etc.), μs-timers, and multiple general-purpose input-output (GPIO) pins suitable for running simple, time-precise programs to control custom-built electronics.

Single board computers

Inexpensive single-board computers capable of running a mature operating system with graphical-user interface, such as Linux. Like microcontroller boards, they offer a variety of hardware interfaces and GPIO pins to control custom-built electronics.It therefore comes as no surprise that these technologies are also routinely used by research scientists and, especially, educators aiming to customize existing lab equipment or even build sophisticated lab equipment from scratch for a mere fraction of what commercial alternatives cost [6]. Designs for such “Open Labware” include simple mechanical adaptors [7], micropipettes (Fig. 1B) [8], and an egg-whisk–based centrifuge [9] as well as more sophisticated equipment such as an extracellular amplifier for neurophysiological experiments [10], a thermocycler for PCR [11], or a two-photon microscope [12]. At the same time, conceptually related approaches are also being pursued in chemistry [1315] and material sciences [16,17]. See also AreaProjectSourceMicroscopySmartphone Microscope http://www.instructables.com/id/10-Smartphone-to-digital-microscope-conversion iPad Microscope http://www.thingiverse.com/thing:31632 Raspberry Pi Microscope http://www.thingiverse.com/thing:385308 Foldscope http://www.foldscope.com/ Molecular BiologyThermocycler (PCR) http://openpcr.org/ Water bath http://blog.labfab.cc/?p=47 Centrifuge http://www.thingiverse.com/thing:151406 Dremelfuge http://www.thingiverse.com/thing:1483 Colorometer http://www.thingiverse.com/thing:73910 Micropipette http://www.thingiverse.com/thing:255519 Gel Comb http://www.thingiverse.com/thing:352873 Hot Plate http://www.instructables.com/id/Programmable-Temperature-Controller-Hot-Plate/ Magnetic Stirrer http://www.instructables.com/id/How-to-Build-a-Magnetic-Stirrer/ ElectrophysiologyWaveform Generator http://www.instructables.com/id/Arduino-Waveform-Generator/ Open EEG https://www.olimex.com/Products/EEG/OpenEEG/ Mobile ECG http://mobilecg.hu/ Extracellular amplifier https://backyardbrains.com/products/spikerBox Micromanipulator http://www.thingiverse.com/thing:239105 Open Ephys http://open-ephys.org/ OtherSyringe pump http://www.thingiverse.com/thing:210756 Translational Stage http://www.thingiverse.com/thing:144838 Vacuum pump http://www.instructables.com/id/The-simplest-vacuum-pump-in-the-world/ Skinner Box http://www.kscottz.com/open-skinner-box-pycon-2014/ Open in a separate windowSee also S1 Data.  相似文献   

18.
The interplay of lipid acyl hydrolases in inducible plant defense     
Etienne Grienenberger  Pierrette Geoffroy  Jérome Mutterer  Michel Legrand  Thierry Heitz 《Plant signaling & behavior》2010,5(10):1181-1186
  相似文献   

19.
A Review of Principal Studies on the Development and Treatment of Epithelial Ovarian Cancer in the Laying Hen Gallus gallus     
Purab Pal  Kara Nicole Starkweather  Karen Held Hales  Dale Buchanan Hales 《Comparative medicine》2021,71(4):271
Often referred to as the silent killer, ovarian cancer is the most lethal gynecologic malignancy. This disease rarely shows any physical symptoms until late stages and no known biomarkers are available for early detection. Because ovarian cancer is rarely detected early, the physiology behind the initiation, progression, treatment, and prevention of this disease remains largely unclear. Over the past 2 decades, the laying hen has emerged as a model that naturally develops epithelial ovarian cancer that is both pathologically and histologically similar to that of the human form of the disease. Different molecular signatures found in human ovarian cancer have also been identified in chicken ovarian cancer including increased CA125 and elevated E-cadherin expression, among others. Chemoprevention studies conducted in this model have shown that decreased ovulation and inflammation are associated with decreased incidence of ovarian cancer development. The purpose of this article is to review the major studies performed in laying hen model of ovarian cancer and discuss how these studies shape our current understanding of the pathophysiology, prevention and treatment of epithelial ovarian cancer.

Ovarian cancer is the leading cause of death among female gynecologic malignancies, with a 47% 5 y relative survival rate.154 Early detection of the disease is necessary for decreasing the high mortality rate. However, early detection is difficult due to the lack of known specific biomarkers and clinically detectable symptoms until the tumor reaches at an advanced stage. The disease has multiple subtypes. Epithelial ovarian cancer (EOC) is the most common type of ovarian cancer, accounting for about 90% of all reported cases.127,164 EOC is commonly subdivided into 5 histotypes: high-grade serous (HGSOC), low-grade serous, mucinous, endometroid (EC), and clear cell. The histotypes differ in terms of tumor cell morphology, severity, systemic effect, and response to treatment. Among the different subtypes, HGSOC accounts for about 70% of cases of EOC observed in women. HGSOC has a higher mitotic index and is a more aggressive form of cancer with a worse prognosis. HGSOC and low-grade serous histotypes exhibit distinctly different presentations of the disease82,166 and demand different treatment modalities. EC (10% to 20%), mucinous (5% to 20%), and clear cell (3% to 10%) histotypes are less common forms of the disease. The subtypes of EOC also differ in terms of 5 y survival rates of patients; that is, HGSOC (20% to 35%), EC (40% to 63%), mucinous (40% to 69%), and clear cell (35% to 50%).20,76,148Developing a representative animal model for EOC has been challenging due to the histologic and pathologic differences among different subtypes of EOC. While developing a reliable animal model is challenging due to the vast complexity and limited understanding of the origin of the disease, laying hens naturally develop EOC that is histopathologically very similar to the human form of the disease (Figure 1).15 All the different human ovarian cancer histotypes have been observed in laying hen ovarian cancer (Figure 2). In addition, the presentation of the disease in chickens is remarkably similar to the human form of the disease, with early-stage ovarian cancer in laying hens having similar precursor lesions as occur in women.15 The laying hen develops ovarian cancer spontaneously, allowing analysis of early events and investigation into the natural course of the disease, as tumors can be examined as they progress from normal to late-stage ovarian carcinoma. The gross appearance of these stages is shown in Figure 3.Open in a separate windowFigure 1.Gross pathologic presentation of chicken compared with human ovarian cancer. The remarkably similar presentation in hens (A,B) and women (C,D) at the gross anatomic level with profuse abdominal ascites and peritoneal dissemination of metastasis. A) Ascites in abdominal cavity chicken with advanced ovarian cancer (photo credit: DB Hales); (B) Chicken ovarian cancer with extensive peritoneal dissemination of metastasis (photo credit: DB Hales); (C) Distended abdomen from ascites fluid accumulation in woman with ovarian cancer (http://www.pathguy.com/bryanlee/ovca.html) (D) Human ovarian cancer with extensive peritoneal dissemination of metastasis (http://www.pathguy.com/bryanlee/ovca.html).Open in a separate windowFigure 2.Gross anatomic appearance of different stages of ovarian cancer in the chicken The progression from the normal hen ovary to late-stage metastatic ovarian cancer. (A) Normal chicken ovary showing hierarchal clutch of developing follicles and postovulatory follicle; (B) Stage 1 ovarian cancer, confined to ovary with vascularized follicles; (C) Stage 2/3 ovarian cancer, metastasis locally to peritoneal cavity with ascites; (D) Stage 4 ovarian cancer, late stage with metastasis to lung and liver with extensive ascites (photo credits: DB Hales).Open in a separate windowFigure 3.Histologic subtypes in chicken compared with human ovarian cancers. H and E staining of formalin fixed paraffin embedded tissues from hens with ovarian cancer (A through D) and women (E through G). (A) Chicken clear cell carcinoma; (B) Chicken endometrioid carcinoma; (C) Chicken mucinous adenocarcinoma; (D) Chicken serous papillary adenocarcinoma (photo credits: DB Hales). (E) Human clear cell carcinoma; (F) Human endometrioid carcinoma; (G) Human mucinous cystadenocarcinoma; (H) Human serous adenocarcinoma (https://www.womenshealthsection.com).Over the past 2 decades, the laying hen has emerged as a valuable experimental model for EOC, in addition to other in vivo models such as Patient-Derived Xenografts (PDX) and Genetically Engineered Mouse Models (GEMMs). Comparison of the hen model with other animal models has been reviewed elsewhere.72 Modern-day laying hens, such as the white leghorn, have been selected from their ancestor red jungle fowl57 for decreased broodiness and persistent ovulation, resulting in approximately one egg per day, if proper nutrition and light-dark cycles are maintained. Daily rupture and consequent repair of the ovarian surface epithelia (OSE) due to the persistent ovulation promotes potential error during rapid DNA replication. This increases the probability of oncogenic mutations, ultimately leading to neoplasia.137 Inflammation resulting from continuous ovulation also promotes the natural development of EOC.81 By the age of 2.5 to 3 y, laying hens have undergone a similar number of ovulations as a perimenopausal woman. The risk of ovarian cancer in white leghorn hens in this time (4%) is similar to the lifetime risk of ovarian cancer in women (0.35% to 8.8%).125 By the age of 4 to 6 y, the risk of ovarian cancer in hens rises to 30% to 60%.54 The incidence of ovarian carcinoma in the hens, however, depends on the age, genetic strain,80 and the egg-laying frequency of the specific breed.54 The common white leghorn hen has routinely been employed in chicken ovarian cancer studies. On average, hens are exposed to 17 h of light per day, with lights turned on at 0500 h and turned off at 2200 h. The laying hen model of EOC does present some considerable challenges. Despite its great utility for research, the model is still used mainly by agricultural poultry scientists and a small number of ovarian cancer researchers.Comprehensive and proper vivarium support is required to conduct large-scale cancer prevention studies. Only a few facilities are available for biomedical chicken research, including University of Illinois Urbana-Champaign, Cornell University, Penn State University, NC State, Auburn University, and MS State University. Another difficulty is a lack of available antibodies specific for chicken antigens. Because of the structural dissimilarities between most human proteins and murine antigens to their chicken counterparts, cross-reactivity of available antibodies is also limited. The entire chicken genome was sequenced in 2004;78 however, the chromosomal locus of many key genes, such as p53, are still unknown. Overall, humans and chickens share about 60% of genetic commonality, whereas humans and rats share about 88% of their genes. Specific pathway-mutated strains of chickens are not yet available, limiting the ability to study key pathways in carcinogenesis and prevention of cancer using this model. Although all 5 different subtypes of ovarian cancer are present in hens, their most predominant subtype is different from women. Close to 70% of women diagnosed with ovarian cancer have serous EOC, while the predominant subtype reported in hens is endometrioid.15 However, these comparisons are complicated because observations of cancer in hens consist of both early and late stages of the disease, wherein women, most of the data is from late stage and aggressive ovarian carcinoma.The spontaneous onset of ovarian cancer and the histologic and pathologic similarities to the human form of the disease make laying hens an excellent model for continued research on EOC. To date, a large number of studies have been performed on laying hens. Here we have divided the current studies into 2 groups— (A) studies that have described the molecular presentation of EOC to be similar to that in women; (AuthorYearSignificanceKey molecular targetsCitationHaritani and colleagues.1984Investigating ovarian tumors for key gene signaturesOvalbumin 71 Rodriguez-Burford and colleagues.2001Investigating expressions of clinically important prognostic markers in cancerous hensCA125, cytokeratin AE1/AE3, pan cytokeratin, Lewis Y, CEA, Tag 72, PCNA, EGFR, erbB-2, p27, TGF{α}, Ki-67, MUC1, and MUC2 135 Giles and colleagues.2004, 2006Investigating ovarian tumors for key gene signaturesOvalbumin, PR, PCNA, Vimentin62, 63Jackson and colleagues.2007CA125 expression in hen ovarian tumorsCA125 79 Stammer and colleagues.2008SELENBP1 downregulation in hen ovarian tumorsSELENBP1 149 Hales and colleagues.2008Cyclooxygenase expressions in hen ovarian tumorsCOX1, COX2, PGE2 67 Urick and colleagues.2008-2009VEGF expression in cultured ascites cells from hen ovarian tumorsVEGF160, 161Ansenberger and colleagues.2009Elevation of E-cadherin in hen ovarian tumorsE-cad 6 Hakim and colleagues.2009Investigating oncogenic mutations in hen ovarian tumorsp53, K-ras, H-ras 66 Zhuge and colleagues.2009CYP1B1 levels in chicken ovarian tumorsCYP1B1 175 Seo and colleagues.2010Upregulation of Claudin-10 in hen ovarian tumorsClaudin-10 145 Trevino and colleagues.2010Investigating ovarian tumors for key gene signaturesOvalbumin, Pax2, SerpinB3, OVM, LTF, RD 157 Choi and colleagues.2011Upregulation of MMP-3 in hen ovarian tumor stromaMMP-3 28 Barua and colleagues.2012Upregulation of DR6 in hen ovarian tumorsDR6 16 Lee and colleagues.2012-2014Upregulation of DNA methylation in hen ovarian tumorsDNMT1, DNMT3A, DNMT3B,
SPP1, SERPINB11, SERPINB1394, 101, 103, 104Lim and colleagues.2013-2014Key genes upregulated in endometrioid hen tumorsAvBD-11, CTNNB1, Wnt4102, 11, 100Bradaric and colleagues.2013Investigating immune cells in hen ovarian tumors 23 Ma and colleagues.2014Identifying unique proteins from proteomic profilingF2 thrombin, ITIH2 106 Hales and colleagues.2014Key genes upregulated in hen ovarian tumorsPAX2, MSX2, FOXA2, EN1 68 Parada and colleagues,2017Unique ganglioside expressed in hen ovarian tumorsNeuGcGM3 124 Open in a separate windowTable 2.Ovarian cancer prevention studies using laying hen model
AuthorYearSignificanceCitation
Barnes and colleagues.2002Medroxyprogesterone study 14
Johnson and colleagues.2006Different genetic strain of laying hens (C strain and K strain) 80
Urick and colleagues.2009Dietary aspirin in laying hens 161
Giles and colleagues.2010Restricted Ovulator strain 61
Carver and colleagues.2011Calorie-restricted hens 25
Eilati and colleagues.2012-2013Dietary flaxseed in laying hens43, 44, 45
Trevino and colleagues.2012Oral contraceptives in laying hens 156
Rodriguez and colleagues.2013Calorie-restricted hens with or without Vitamin D and progestin 136
Mocka and colleagues.2017p53 stabilizer CP-31398 in laying hens 112
Open in a separate window  相似文献   

20.
Ion channel regulation by protein S-acylation     
Michael J. Shipston 《The Journal of general physiology》2014,143(6):659-678
Protein S-acylation, the reversible covalent fatty-acid modification of cysteine residues, has emerged as a dynamic posttranslational modification (PTM) that controls the diversity, life cycle, and physiological function of numerous ligand- and voltage-gated ion channels. S-acylation is enzymatically mediated by a diverse family of acyltransferases (zDHHCs) and is reversed by acylthioesterases. However, for most ion channels, the dynamics and subcellular localization at which S-acylation and deacylation cycles occur are not known. S-acylation can control the two fundamental determinants of ion channel function: (1) the number of channels resident in a membrane and (2) the activity of the channel at the membrane. It controls the former by regulating channel trafficking and the latter by controlling channel kinetics and modulation by other PTMs. Ion channel function may be modulated by S-acylation of both pore-forming and regulatory subunits as well as through control of adapter, signaling, and scaffolding proteins in ion channel complexes. Importantly, cross-talk of S-acylation with other PTMs of both cysteine residues by themselves and neighboring sites of phosphorylation is an emerging concept in the control of ion channel physiology. In this review, I discuss the fundamentals of protein S-acylation and the tools available to investigate ion channel S-acylation. The mechanisms and role of S-acylation in controlling diverse stages of the ion channel life cycle and its effect on ion channel function are highlighted. Finally, I discuss future goals and challenges for the field to understand both the mechanistic basis for S-acylation control of ion channels and the functional consequence and implications for understanding the physiological function of ion channel S-acylation in health and disease.Ion channels are modified by the attachment to the channel protein of a wide array of small signaling molecules. These include phosphate groups (phosphorylation), ubiquitin (ubiquitination), small ubiquitin-like modifier (SUMO) proteins (SUMOylation), and various lipids (lipidation). Such PTMs are critical for controlling the physiological function of ion channels through regulation of the number of ion channels resident in the (plasma) membrane; their activity, kinetics, and modulation by other PTMs; or their interaction with other proteins. S-acylation is one of a group of covalent lipid modifications (Resh, 2013). However, unlike N-myristoylation and prenylation (which includes farnesylation and geranylgeranylation), S-acylation is reversible (Fig. 1). Because of the labile thioester bond, S-acylation thus represents a dynamic lipid modification to spatiotemporally control protein function. The most common form of S-acylation, the attachment of the C16 lipid palmitate to proteins (referred to as S-palmitoylation), was first described more than 30 years ago in the transmembrane glycoprotein of the vesicular stomatitis virus and various mammalian membrane proteins (Schmidt and Schlesinger, 1979; Schlesinger et al., 1980). A decade later, S-acylated ion channels—rodent voltage-gated sodium channels (Schmidt and Catterall, 1987) and the M2 ion channel from the influenza virus (Sugrue et al., 1990)—were first characterized. Since then, more than 50 distinct ion channel subunits have been experimentally demonstrated to be S-acylated (El-Husseini and Bredt, 2002; Linder and Deschenes, 2007; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Resh, 2012). In the last few years, with the cloning of enzymes controlling S-acylation and development of various proteomic tools, we have begun to gain substantial mechanistic and physiological insight into how S-acylation may control multiple facets of the life cycle of ion channels: from their assembly, through their trafficking and regulation at the plasma membrane, to their final degradation (Fig. 2).Open in a separate windowFigure 1.Protein S-acylation: a reversible lipid posttranslational modification of proteins. (A) Major lipid modifications of proteins. S-acylation is reversible due to the labile thioester bond between the lipid (typically, but not exclusively, palmitate) and the cysteine amino acid of is target protein. Other lipid modifications result from stable bond formation between either the N-terminal amino acid (amide) or the amino acid side chain in the protein (thioether and oxyester). The zDHHC family of palmitoyl acyltransferases mediates S-acylation with other enzyme families controlling other lipid modifications: N-methyltransferase (NMT) controls myristoylation of many proteins such as the src family kinase, Fyn kinase; and amide-linked palmitoylation of the secreted sonic hedgehog protein is mediated by Hedgehog acyltransferase (Hhat), a membrane-bound O-acyl transferase (MBOAT) family. Prenyl transferases catalyze farnesyl (farnesyltransferase, FTase) or geranylgeranyl (geranylgeranyl transferase I [GGTase I] and geranylgeranyl transferase II [GGTase II]) in small GTPase proteins such as RAS and the Rab proteins, respectively. Porcupine (Porcn) is a member of the MBOAT family acylates secreted proteins such as Wnt. (B) zDHHC enzymes typically use coenzyme A (CoA)-palmitate; however, other long chain fatty acids (either saturated or desaturated) can also be used. Deacylation is mediated by several acylthioesterases of the serine hydrolase family. (C) zDHHC acyltransferases (23 in humans) are predicted transmembrane proteins (typically with 4 or 6 transmembrane domains) with the catalytic DHHC domain located in a cytosolic loop.

Table 1.

Pore-forming subunits of ion channels experimentally determined to be S-acylated
ChannelSubunitGeneCandidate S-acylation sitesUniProt IDReferences
Ligand-gated
AMPAGluA1Gria1593FSLGAFMQQGCDISPRSLSGRIP23818Hayashi et al., 2005
819LAMLVALIEFCYKSRSESKRMKP23818Hayashi et al., 2005
GluA2Gria2600FSLGAFMRQGCDISPRSLSGRIP23819Hayashi et al., 2005
826LAMLVALIEFCYKSRAEAKRMKP23819Hayashi et al., 2005
GluA3Gria3605FSLGAFMQQGCDISPRSLSGRIQ9Z2W9Hayashi et al., 2005
831LAMMVALIEFCYKSRAESKRMKQ9Z2W9Hayashi et al., 2005
GluA4Gria4601FSLGAFMQQGCDISPRSLSGRIQ9Z2W8Hayashi et al., 2005
827LAMLVALIEFCYKSRAEAKRMKQ9Z2W8Hayashi et al., 2005
GABAAγ2Gabrg2405QERDEEYGYECLDGKDCASFFCCFEDCRTGAWRHGRIP22723Rathenberg et al., 2004; Fang et al., 2006
KainateGluK2Grik2848KNAQLEKRSFCSAMVEELRMSLKCQRRLKHKPQAPVP39087Pickering et al., 1995
nAChRα4Chrna4263TVLVFYLPSECGEKVTLCISVO70174Alexander et al., 2010; Amici et al., 2012
α7Chrna7NDAlexander et al., 2010; Drisdel et al., 2004
β2Chrnb2NDAlexander et al., 2010
NMDAGluN2AGrin2a838EHLFYWKLRFCFTGVCSDRPGLLFSISRGIYSCIHGVHIEEKKP35436Hayashi et al., 2009
1204SDRYRQNSTHCRSCLSNLPTYSGHFTMRSPFKCDACLRMGNLYDIDP35436Hayashi et al., 2009
GluN2BGrin2b839EHLFYWQFRHCFMGVCSGKPGMVFSISRGIYSCIHGVAIEERQQ01097Hayashi et al., 2009
1205DWEDRSGGNFCRSCPSKLHNYSSTVAGQNSGRQACIRCEACKKAGNLYDISQ01097Hayashi et al., 2009
P2X7P2X7P2rx7361AFCRSGVYPYCKCCEPCTVNEYYYRKKQ9Z1M0Gonnord et al., 2009
469APKSGDSPSWCQCGNCLPSRLPEQRRQ9Z1M0Gonnord et al., 2009
488PEQRRALEELCCRRKPGRCITTQ9Z1M0Gonnord et al., 2009
562DMADFAILPSCCRWRIRKEFPKQ9Z1M0Gonnord et al., 2009
Voltage gated
Potassium
BK, maxiKKCa1.1Kcnma143WRTLKYLWTVCCHCGGKTKEAQKIQ08460Jeffries et al., 2010
635MSIYKRMRRACCFDCGRSERDCSCMQ08460Tian et al., 2008; 2010
Kv1.1Kcna1233SFELVVRFFACPSKTDFFKNIP16388Gubitosi-Klug et al., 2005
Kv1.5Kcna516LRGGGEAGASCVQSPRGECGCQ61762Jindal et al., 2008
583VDLRRSLYALCLDTSRETDL-stopQ61762Zhang et al., 2007; Jindal et al., 2008
SodiumNaV1.2Scn2a1NDSchmidt and Catterall, 1987
640MNGKMHSAVDCNGVVSLVGGPP04775Bosmans et al., 2011
1042LEDLNNKKDSCISNHTTIEIGP04775Bosmans et al., 2011
1172TEDCVRKFKCCQISIEEGKGKP04775Bosmans et al., 2011
Other channels
AquaporinAQP4Aqp43DRAAARRWGKCGHSCSRESIMVAFKP55088Crane and Verkman, 2009; Suzuki et al., 2008
CFTRCFTRCFTR514EYRYRSVIKACQLEEDISKFAEKDP13569McClure et al., 2012
1385RRTLKQAFADCTVILCEHRIEAP13569McClure et al., 2012
ConnexinCx32Gjb1270GAGLAEKSDRCSAC-stopP28230Locke et al., 2006
ENaCENaC βScnn1b33TNTHGPKRIICEGPKKKAMWFLQ9WU38Mueller et al., 2010
547WITIIKLVASCKGLRRRRPQAPYQ9WU38Mueller et al., 2010
ENaC γScnn1g23PTIKDLMHWYCLNTNTHGCRRIVVSRGRLQ9WU39Mukherjee et al., 2014
Influenza M2M240LWILDRLFFKCIYRFFEHGLKQ20MD5Sugrue et al., 1990; Holsinger et al., 1995; Veit et al., 1991
RyR1RYR1Ryr114LRTDDEVVLQCSATVLKEQLKLCLAAEGFGNRLP11716Chaube et al., 2014
110RHAHSRMYLSCLTTSRSMTDKP11716Chaube et al., 2014
243RLVYYEGGAVCTHARSLWRLEP11716Chaube et al., 2014
295EDQGLVVVDACKAHTKATSFCP11716Chaube et al., 2014
527ASLIRGNRANCALFSTNLDWVP11716Chaube et al., 2014
1030ATKRSNRDSLCQAVRTLLGYGP11716Chaube et al., 2014
1664SHTLRLYRAVCALGNNRVAHAP11716Chaube et al., 2014
2011HFKDEADEEDCPLPEDIRQDLP11716Chaube et al., 2014
2227KMVTSCCRFLCYFCRISRQNQP11716Chaube et al., 2014
2316KGYPDIGWNPCGGERYLDFLRP11716Chaube et al., 2014
2353VVRLLIRKPECFGPALRGEGGP11716Chaube et al., 2014
2545EMALALNRYLCLAVLPLITKCAPLFAGTEHRP11716Chaube et al., 2014
3160DVQVSCYRTLCSIYSLGTTKNTYVEKLRPALGECLARLAAAMPVP11716Chaube et al., 2014
3392LLVRDEFSVLCRDLYALYPLLP11716Chaube et al., 2014
3625SKQRRRAVVACFRMTPLYNLPP11716Chaube et al., 2014
Open in a separate windowCommon channel abbreviation and subunit as well as gene names are given. Candidate S-acylation sites: experimentally determined cysteine residues (bold) with flanking 10 amino acids. Underlines indicate predicted transmembrane domains. Amino acid numbering corresponds to the UniProt ID. References: selected original supporting citations.Open in a separate windowFigure 2.Protein S-acylation and regulation of the ion channel lifecycle zDHHCs are found in multiple membrane compartments and regulate multiple steps in the ion channel lifecycle including: (1) assembly and (2) ER exit; (3) maturation and Golgi exit; (4) sorting and trafficking; (5) trafficking and insertion into target membrane; (6) clustering and localization in membrane microdomains; control of properties, activity (7), and regulation by other signaling pathways; and (8) internalization, recycling, and final degradation.

Table 3.

Other channels identified in mammalian palmitoylome screens
ChannelGene
Anion
Chloride channel 6Clcn6
Chloride intracellular channel 1Clic1
Chloride intracellular channel 4Clic4
Tweety homologue 1Ttyh1
Tweety homologue 3Ttyh3
Voltage-dependent anion channel 1Vdac1
Voltage-dependent anion channel 2Vdac2
Voltage-dependent anion channel 3Vdac3
Calcium
Voltage-dependent, L-type subunit α 1SCacna1s
Voltage-dependent, gamma subunit 8Cacng8
Cation
Amiloride-sensitive cation channel 2Accn2
Glutamate
Ionotropic, Δ1Grid1
Perforin
Perforin 1Prf1
Potassium
Voltage-gated channel, subfamily Q, member 2Kcnq2
Sodium
Voltage-gated, type I, αScn1a
Voltage-gated, type III, αScn3a
Voltage-gated, type IX, αScn9a
Transient receptor potential
Cation channel, subfamily V, member 2Trpv2
Cation channel, subfamily M, member 7Trpm7
Open in a separate windowChannels identified in global S-acylation screens (Wan et al., 2007, 2013; Kang et al., 2008; Martin and Cravatt, 2009; Yang et al., 2010; Yount et al., 2010; Merrick et al., 2011; Wilson et al., 2011; Jones et al., 2012; Ren et al., 2013; Chaube et al., 2014) and not independently characterized as in and2.2. Common channel abbreviation and gene names are given.Here, I provide a primer on the fundamentals of S-acylation, in the context of ion channel regulation, along with a brief overview of tools available to interrogate ion channel S-acylation. I will discuss key examples of how S-acylation controls distinct stages of the ion channel life cycle before highlighting some of the key challenges for the field in the future.

Fundamentals of S-acylation: The what, when, where, and how

S-acylation: A fatty modification that controls multiple aspects of protein function.

Protein S-acylation results from the attachment of a fatty acid to intracellular cysteine residues of proteins via a labile, thioester linkage (Fig. 1, A and B). Because the thioester bond is subject to nucleophilic attack, S-acylation, unlike other lipid modifications such as N-myristoylation and prenylation, is reversible. However, for most ion channels, as for other S-acylated proteins, the dynamics of S-acylation are poorly understood. Distinct classes of proteins can undergo cycles of acylation and deacylation that are very rapid (e.g., on the timescale of seconds, as exemplified by rat sarcoma [RAS] proteins), much longer (hours), or essentially irreversible during the lifespan of the protein (El-Husseini and Bredt, 2002; Linder and Deschenes, 2007; Zeidman et al., 2009; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Resh, 2012). For most ion channels, in fact most S-acylated proteins, the identity of the native lipid species attached to specific cysteine residues is also largely unknown. However, the saturated C16:0 lipid palmitate is commonly thought to be the major lipid species in many S-acylated proteins (Fig. 1). Indeed, much of the earliest work on S-acylation involved the metabolic labeling of proteins in cells with tritiated [3H]palmitate, an approach that still remains useful and important. However, lipids with different chain lengths and degrees of unsaturation (such as oleic and stearic acids) can also be added to cysteines via a thioester linkage, potentially allowing differential control of protein properties through the attachment of distinct fatty acids (El-Husseini and Bredt, 2002; Linder and Deschenes, 2007; Zeidman et al., 2009; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Resh, 2012).S-acylation increases protein hydrophobicity and has thus been implicated in controlling protein function in many different ways. Most commonly, as with membrane-associated proteins like RAS and postsynaptic density protein 95 (PSD-95), S-acylation controls membrane attachment and intracellular trafficking. However, S-acylation can also control protein–protein interactions, protein targeting to membrane subdomains, protein stability, and regulation by other PTMs such as phosphorylation (El-Husseini and Bredt, 2002; Fukata and Fukata, 2010; Linder and Deschenes, 2007; Greaves and Chamberlain, 2011; Shipston, 2011; Resh, 2012). Evidence for all these mechanisms in controlling ion channel function is beginning to emerge.

Enzymatic control of S-acylation by zinc finger–containing acyltransferase (zDHHC) transmembrane acyltransferases.

Although autoacylation of some proteins has been reported in the presence of acyl coenzyme A (acyl-CoA; Linder and Deschenes, 2007), most cellular S-acylation, in organisms from yeast to humans, is thought to be enzymatically driven by a family of protein acyltransferases (gene family: zDHHC, with ∼23 members in mammals). These acyltransferases are predicted to be transmembrane zinc finger containing proteins (Fig. 1 C) that include a conserved Asp-His-His-Cys (DHHC) signature sequence within a cysteine-rich stretch of ∼50 amino acids critical for catalytic activity (Fukata et al., 2004). Although the enzymatic activity and lipid specificity of all of the zDHHC family proteins has not been elucidated, S-acylation is thought to proceed through a common, two step “ping pong” process (Mitchell et al., 2010; Jennings and Linder, 2012). However, different zDHHC enzymes may show different acyl-CoA substrate specificities. For example, zDHHC3 activity is reduced by acyl chains of >16 carbons (e.g., stearoyl CoA), whereas zDHHC2 efficiently transfers acyl chains of 14 carbons or longer (Jennings and Linder, 2012). The local availability of different acyl-CoA species may thus play an important role in differentially controlling protein S-acylation.We know very little about how zDHHC activity and function are regulated. Dimerization of zDHHCs 2 and 3 reduces their zDHHC activity compared with the monomeric form (Lai and Linder, 2013). Moreover, zDHHCs undergo autoacylation and contain predicted sites for other posttranslational modifications. Almost half of all mammalian zDHHCs contain a C-terminal PSD-95, Discs large, and ZO-1 (PDZ) domain binding motif, allowing them to assemble with various PDZ domain proteins that regulate ion channels (such as GRIP1b and PSD-95; Thomas and Hayashi, 2013). Other protein interaction domains are also observed in zDHHCs, such as ankyrin repeats in zDHHC17 and zDHHC13 (Greaves and Chamberlain, 2011). Indeed, increasing evidence suggests that various ion channels—including the ligand-gated γ-aminobutyric (GABAA), α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA), and NMDA receptors and the large conductance calcium- and voltage-activated (BK) potassium channels—can assemble in complexes with their cognate zDHHCs.The expansion of the number of zDHHCs in mammals (23 vs. 7 in yeast), together with increased prevalence of PDZ interaction motifs, likely represents evolutionary gain-of-function mechanisms to diversify zDHHC function (Thomas and Hayashi, 2013). Evolutionary gain of function is also seen in ion channel subunit orthologues through acquisition of S-acylated cysteine residues absent in orthologues lower in the phylogenetic tree (such as the transmembrane domain 4 [TM4] sites in GluA1–4 subunits of AMPA receptors [Thomas and Hayashi, 2013] and the sites in the alternatively spliced stress-regulated exon [STREX] insert in the C terminus of the BK channel [Tian et al., 2008]). Importantly, some zDHHCs may have additional roles beyond their acyltransferase function. For example, the Drosophila melanogaster zDHHC23 orthologue lacks the catalytic DHHC sequence, and thus protein acyltransferase activity, and is a chaperone involved in protein trafficking (Johswich et al., 2009), whereas mammalian zDHHC 23 has a functional zDHHC motif and, in addition to S-acylating BK channels (Tian et al., 2012), can bind and regulate, but does not S-acylate, neuronal nitric oxide synthase (nNOS; Saitoh et al., 2004).However, as with most S-acylated proteins, the identity of the zDHHCs that modify specific cysteine residues on individual ion channels is not known. Indeed, relatively few studies have tried to systematically identify the zDHHCs controlling ion channel function (Tian et al., 2010, 2012). Thus we are largely ignorant of the extent to which different zDHHCs may have specific ion channel targets or may display specificity. Some details are beginning to emerge: for example, zDHHC3 appears to be a rather promiscuous acyltransferase reported to S-acylate several ion channels (Keller et al., 2004; Hayashi et al., 2005, 2009; Tian et al., 2010), whereas distinct sites on the same ion channel subunit can be modified by distinct subsets of zDHHCs (Tian et al., 2010, 2012). Although we are still in the foothills of understanding the substrates and physiological roles of different zDHHCs, mutation or loss of function in zDHHCs is associated with an increasing number of human disorders, including cancers, various neurological disorders (such as Huntington’s disease and X-linked mental retardations), and disruption of endocrine function in diabetes (Linder and Deschenes, 2007; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Resh, 2012).

Deacylation is controlled by acylthioesterases.

Protein deacylation is enzymatically driven by a family of acylthioesterases that belong to the serine hydrolase superfamily (Zeidman et al., 2009; Bachovchin et al., 2010). Indeed, using a broad spectrum serine lipase inhibitor, global proteomic S-acylation profiling identified a subset of serine hydrolases responsible for depalmitoylation (Martin et al., 2012). This study identified both the previously known acylthioesterases as well as potential novel candidate acylthioesterases. The acylthioesterases responsible for deacylating ion channels, as for most other acylated membrane proteins, have not been clearly defined. Furthermore, the extent to which different members of the serine hydrolase superfamily display acylthioesterase activity toward ion channels is not known. Moreover, whether additional mechanisms of nucleophilic attack of the labile thioester bond may also mediate deacylation is not known.Homeostatic control of deacylation of many signaling proteins is likely affected by a family of cytosolic acyl protein thioesterases including lysophospholipase 1 (LYPLA1; Yeh et al., 1999; Devedjiev et al., 2000) and lysophospholipase 2 (LYPLA2; Tomatis et al., 2010). These enzymes show some selectivity for different S-acylated peptides (Tomatis et al., 2010). Indeed, LYPLA1, but not LYPLA2, deacylates the S0-S1 loop of BK channels, leading to Golgi retention of the channel (Tian et al., 2012). A splice variant of the related LYPLAL1 acylthioesterases can also deacylate the BK channel S0-S1 loop, although the crystal structure of LYPLAL1 suggests it is likely to have a preference for lipids with shorter chains than palmitate (Bürger et al., 2012). Thus, whether lipid preference depends on protein interactions or if BK channels have multiple lipid species at the multicysteine S0-S1 site remain unknown. Relatively little is known about the regulation of these acylthioesterases; however, both LYPLA1 and LYPLA2 are themselves S-acylated. This controls their trafficking and association with membranes (Kong et al., 2013; Vartak et al., 2014) and may be important for accessing the thioesterase bond at the membrane interface. Additional mechanisms may promote accessibility of thioesterases to target cysteines. For example, the prolyl isomerase protein FKBP12 binds to palmitoylated RAS, and promotes RAS deacylation via a proline residue near the S-acylated cysteine (Ahearn et al., 2011).Upon lysosomal degradation, many proteins are deacylated by the lysosomal palmitoyl protein thioesterase (PPT1; Verkruyse and Hofmann, 1996), and mutations in PPT1 lead to the devastating condition of infantile neuronal ceroid lipofuscinosis (Vesa et al., 1995; Sarkar et al., 2013). However, PPT1 can also be found in synaptic and other transport vesicles, and genetic deletion of PPT1 in mice may have different effects on similar proteins, which suggests roles beyond just lysosomal mediated degradation. For example, in PPT1 knockout mice the total expression and surface membrane abundance of the GluA4 AMPA receptor subunit was decreased, whereas PPT1 knockout had no effect on GluA1 or GluA2 AMPA subunits nor on NMDA receptor subunit expression or surface abundance (Finn et al., 2012).However, for most ion channels, the questions of which enzymes control deacylation, where this occurs in cells, and how the time course of acylation–deacylation cycles are regulated are largely unknown. Thus, whether deacylation plays an active role in channel regulation remains poorly understood.

S-acylation occurs at membrane interfaces.

Because the zDHHCs are transmembrane proteins and the catalytic DHHC domain is located at the cytosolic interface with membranes (Fig. 1 C), S-acylation of ion channels occurs at membrane interfaces. Although overexpression studies of recombinant mammalian zDHHCs in heterologous expression systems have indicated that most zDHHCs are localized to either the endoplasmic reticular or Golgi apparatus membranes (or both; Ohno et al., 2006), some zDHHCs are also found in other compartments, including the plasma membrane and trafficking endosomes (Thomas et al., 2012; Fukata et al., 2013). We know very little about the regulation and subcellular localization of most native zDHHC enzymes in different cell types, in large part because of the lack of high-quality antibodies that recognize native zDHHCs. However, some enzymes, including zDHHC2, can dynamically shuttle between different membrane compartments. Activity-dependent redistribution of zDHHC2 in neurons (Noritake et al., 2009) controls S-acylation of the postsynaptic scaffolding protein PSD-95, thereby regulating NMDA receptor function. Intriguingly, as ion channels themselves determine cellular excitability, this may provide a local feedback mechanism to regulate S-acylation status. Thus, although different zDHHCs may reside in multiple membrane compartments through which ion channels traffic, the subcellular location at which most ion channels are S-acylated, as well as the temporal dynamics, is largely unknown. As discussed below (see the “Tools to analyze ion channel S-acylation” section), we are starting to unravel some of the details, with ER exit, Golgi retention, recycling endosomes, and local plasma membrane compartments being key sites in the control of ion channel S-acylation (Fig. 2).

Local membrane and protein environment determines cysteine S-acylation.

The efficiency of S-acylation of cysteine residues is likely enhanced by its localization at membranes because the local concentration of fatty acyl CoA is increased near hydrophobic environments (Bélanger et al., 2001). Furthermore, S-acylation of polytopic transmembrane proteins such as ion channels would be facilitated when S-acylated cysteines are bought into close proximity of membranes by membrane targeting mechanisms such as transmembrane helices (Figs. 3 and and4).4). However, the S-acylated cysteine is located within 10 amino acids of a transmembrane domain in only ∼20% of identified S-acylated ion channel subunits, such as the TM4 site of GluA1–4 (and2).2). Most S-acylated cysteines are located either within intracellular loops (∼40%: Fig. 3, A and B) or the N- or C-terminal cytosolic domains (∼5% and 35%, respectively; Fig. 3, A and B). Furthermore, the majority of S-acylated cysteines located in intracellular loops or intracellular N- or C-terminal domains of ion channel subunits are within predicted regions of protein disorder (Fig. 3 B). This suggests that S-acylation may provide a signal to promote conformational restraints on such domains, in particular by providing a membrane anchor. For these sites, additional initiating membrane association signals are likely required adjacent to the site of S-acylation. Likely candidates include other hydrophobic domains (as for the TM2 site in GluA1–4 subunits; Fig. 4 A) and other lipid anchors (e.g., myristoylation in src family kinases, such as Fyn kinase). However, in >30% of S-acylated ion channels, the S-acylated cysteine is juxtaposed to a (poly) basic region of amino acids that likely allows electrostatic interaction with negative membrane phospholipids. The BK channel pore-forming α subunit, encoded by the KCNMA1 gene, provides a clear example of this latter mechanism. This channel is S-acylated within an alternatively spliced domain (STREX) in its large intracellular C terminus (Fig. 4 C). Immediately upstream of the S-acylated dicysteine motif is a polybasic region enriched with arginine and lysine. Site-directed mutation of these basic amino acids disrupts S-acylation of the downstream cysteine residues (Jeffries et al., 2012). Furthermore, phosphorylation of a consensus PKA site (i.e., introduction of negatively charged phosphate) into the polybasic domain prevents STREX S-acylation. Thus, at the STREX domain, an electrostatic switch, controlled by phosphorylation, is an important determinant of BK channel S-acylation. In other proteins, cysteine reactivity is also enhanced by proximity to basic (or hydrophobic) residues (Bélanger et al., 2001; Britto et al., 2002; Kümmel et al., 2010). Furthermore, cysteine residues are subject to a range of modifications including nitrosylation, sulphydration, reduction-oxidation (REDOX) modification, and formation of disulphide bonds (Sen and Snyder, 2010). Evidence is beginning to emerge that these reversible modifications are mutually competitive for S-acylation of target cysteines (see the “S-acylation and posttranslational cross-talk controls channel trafficking and activity” section; Ho et al., 2011; Burgoyne et al., 2012).Open in a separate windowFigure 3.S-acylation sites in ion channel pore-forming subunits. (A) Schematic illustrating different locations of cysteine S-acylation in transmembrane ion channels subunits. (B) Relative proportion of identified S-acylated cysteine residues: in each location indicated in A (top); in -C-, -CC-, or -Cx(2–3)C- motifs (middle); or in cytosolic regions of predicted protein disorder (bottom; determined using multiple algorithms on the DisProt server, http://www.disprot.org/metapredictor.php; Sickmeier et al., 2007) for transmembrane ion channel pore-forming subunits.Open in a separate windowFigure 4.Multisite S-acylation in ion channels controls distinct functions. (A–C) Schematic illustrating location of multiple S-acylated domains in AMPA receptor GluA1–4 subunits (A), NMDA receptor GluN2A subunits (B), and BK channel pore-forming α subunits (C), encoded by the Kcnma1 gene. Each domain confers distinct functions/properties on the respective ion channel and is regulated by distinct zDHHCs (see the “Control of ion channel cell surface expression and spatial organization in membranes” section for further details).

Table 2.

Accessory subunits and selected ion channel adapter proteins
ChannelSubunitGeneCandidate S-acylation sitesUniProt IDReferences
Voltage gated
CalciumCaVβ2aCacnb21MQCCGLVHRRRVRVQ8CC27Chien et al., 1996; Stephens et al., 2000; Heneghan et al., 2009; Mitra-Ganguli et al., 2009
PotassiumKChip2Kcnip234LKQRFLKLLPCCGPQALPSVSEQ9JJ69Takimoto et al., 2002
KChip3Kcnip335PRFTRQALMRCCLIKWILSSAAQ9QXT8Takimoto et al., 2002
BK β4Kcnmb4193VGVLIVVLTICAKSLAVKAEAQ9JIN6Chen et al., 2013
Adapter proteins that interact with ion channelsPICK1Pick1404TGPTDKGGSWCDS-stopQ62083Thomas et al., 2013
Grip1bGrip11MPGWKKNIPICLQAEEQEREQ925T6-2Thomas et al., 2012; Yamazaki et al., 2001
psd-95Dlg41MDCLCIVTTKKYRQ62108Topinka and Bredt, 1998
S-delphilinGrid2ip1MSCLGIFIPKKHQ0QWG9-2Matsuda et al., 2006
Ankyrin-GAnk360YIKNGVDVNICNQNGLNALHLF1LNM3He et al., 2012
Open in a separate windowCommon channel abbreviation and subunit as well as gene names are given. Candidate S-acylation sites: experimentally determined cysteine residues (bold) with flanking 10 amino acids. Underlines indicate predicted transmembrane domains. Amino acid numbering corresponds to the UniProt ID. References: selected original supporting citations.Although these linear amino acid sequence features are likely to be important for efficient S-acylation, there is no canonical “consensus” S-acylation motif analogous to the linear amino acid sequences that predict sites of phosphorylation. Of the experimentally validated ion channel subunits shown to be S-acylated, ∼70% of candidate S-acylated cysteines are predominantly characterized as single cysteine (-C-) motifs, whereas dicysteine motifs (-CC-) and (CX(1–3)C-) motifs comprise ∼10% and 20% of all sites, respectively (Fig. 3 B). However, several freely available online predictive tools have proved successful in characterizing potential new palmitoylation targets. In particular, the latest iteration of the multiplatform CSS-palm 4.0 tool (Ren et al., 2008) exploits a Group-based prediction algorithm by comparing the surrounding amino acid sequence similarity to that of a set of 583 experimentally determined S-acylation sites from 277 distinct proteins. CSS-palm 4.0 predicts >80% of the experimentally identified ion channel S-acylation sites (Location of S-acylated cysteine is important for differential control of channel function.Many proteins are S-acylated at multiple sites. A remarkable example of this, in the ion channel field, is the recent identification of 18 S-acylated cysteine residues in the skeletal muscle ryanodine receptor/Ca2+-release channel (RyR1). The S-acylated cysteine residues are distributed throughout the cytosolic N terminus, including domains important for protein–protein interactions (Chaube et al., 2014). Although deacylation of skeletal muscle RyR1 reduces RyR1 activity, the question of which of these cysteine residues in RyR1 are important for this effect and whether distinct S-acylated cysteines in RyR1 control different functions and/or properties remains to be determined.However, both ligand-gated (NMDA and AMPA) and voltage-gated (BK) channels provide remarkable insights into how S-acylation of different domains within the same polytopic protein can exert fundamentally distinct effects (Fig. 4). For example, S-acylation of the hydrophobic cytosolic TM2 domain located at the membrane interface of the AMPA GluA1 subunit (Fig. 4 A) decreases AMPA receptor surface expression by retaining the subunit at the Golgi apparatus (Hayashi et al., 2005). In contrast, depalmitoylation of the C-terminal cysteine in GluA1 results in enhanced PKC-dependent phosphorylation of neighboring serine residues, which results in increased interaction with the actin-binding protein 4.1N in neurons, leading to enhanced AMPA plasma membrane insertion (Lin et al., 2009). S-acylation of the C-terminal cluster of cysteine residues (Fig. 4 B, Cys II site) in GluN2A and GluN2B controls Golgi retention, whereas palmitoylation of the cysteine cluster (Cys I site) proximal to the M4 transmembrane domain controls channel internalization (Hayashi et al., 2009). Distinct roles of S-acylation on channel trafficking and regulation are also observed in BK channels (Figs. 4 C and and5).5). S-acylation of the N-terminal intracellular S0-S1 linker controls surface expression, in part by controlling ER and Golgi exit of the channel (Jeffries et al., 2010; Tian et al., 2012), whereas S-acylation of the large intracellular C terminus, within the alternatively spliced STREX domain, controls BK channel regulation by AGC family protein kinases (Tian et al., 2008; Zhou et al., 2012).Open in a separate windowFigure 5.S-acylation controls BK channel trafficking and regulation by AGC family protein kinases via distinct sites. The BK channel STREX splice variant pore-forming α subunit is S-acylated at two sites: the S0-S1 loop and the STREX domain in the large intracellular C terminus. S-acylation of the S0-S1 loop promotes high surface membrane expression of the channel; thus, deacylation of this site decreases the number of channels at the cell surface (see the “Control of ion channel cell surface expression and spatial organization in membranes” section for further details). In contrast, S-acylation of the STREX domain allows inhibition of channel activity by PKA-mediated phosphorylation of a PKA serine motif (closed hexagon) immediately upstream of the palmitoylated cysteine residues in STREX. In the S-acylated state, PKC has no effect on channel activity even though a PKC phosphorylation site serine motif is located immediately downstream of the STREX domain (open triangle). Deacylation of STREX dissociates the STREX domain from the plasma membrane, and exposes the PKC serine motif so that it can now be phosphorylated by PKC (closed triangle), resulting in channel inhibition. In the deacylated state, PKA has no effect on channel activity (open hexagon). Thus, deacylation of the STREX domain switches channel regulation from a PKA-inhibited to a PKC-inhibited phenotype (see the “S-acylation and posttranslational cross-talk controls channel trafficking and activity” section for further details).How does S-acylation of distinct domains control such behavior, and are distinct sites on the same protein acylated by distinct zDHHCs? A systematic small interfering RNA (siRNA) screen of zDHHC enzymes mediating BK channel S-acylation indicated that distinct subsets of zDHHCs modify discrete sites. The S0-S1 loop is S-acylated by zDHHCs 22 and 23, whereas the STREX domain is S-acylated by several zDHHCs including 3, 9, and 17 (Tian et al., 2008, 2012). In both cases, each domain has two distinct S-acylated cysteines; however, whether these cysteines are differentially S-acylated by specific zDHHCs is unknown, Furthermore, whether multiple zDHHCs are required because the domains undergo repeated cycles of S-acylation and deacylation, and thus different zDHHCs function at different stages of the protein lifecycle, remains to be determined. Although systematic siRNA screens have, to date, not been performed on other ion channels, data from other multiply S-acylated channels, such as NMDA, AMPA, and BK channel subunits, supports the hypothesis that zDHHCs can show substrate specificity (Hayashi et al., 2005, 2009; Tian et al., 2010).It is generally assumed that S-acylation facilitates the membrane association of protein domains. This is clearly the case for peripheral membrane proteins, such as RAS or PSD-95, but direct experimental evidence for S-acylation controlling membrane association of the cytosolic domains of transmembrane proteins is largely elusive. One of the best examples involves the large C-terminal domain of the BK channel, which comprises more than two-thirds of the pore-forming subunit (Fig. 5). In the absence of S-acylation of the STREX domain, or exclusion of the 59–amino acid STREX insert, the BK channel C terminus is cytosolic (Tian et al., 2008). However, if the STREX domain is S-acylated, the entire C terminus associates with the plasma membrane, a process that can be dynamically regulated by phosphorylation of a serine immediately upstream of the S-acylated cysteines in the STREX domain (Tian et al., 2008). This S-acylation–dependent membrane association markedly affects the properties and regulation of the channel (Jeffries et al., 2012) and has been proposed to confer significant structural rearrangements. In support of such structural rearrangement, S-acylated STREX channels are not inhibited by PKC-dependent phosphorylation even though a PKC phosphorylation site serine motif, conserved in other BK channel variants, is present downstream of the STREX domain. In other BK channel variants lacking the STREX insert, this PKC site is required for channel inhibition by PKC-dependent phosphorylation. However, after deacylation of the STREX domain, PKC can now phosphorylate this PKC phosphorylation serine motif, which suggests that the site has become accessible, consequently resulting in channel inhibition (Fig. 5; Zhou et al., 2012).How might S-acylation of a cysteine residue juxtaposed to another membrane anchoring domain control protein function? The simplest mechanism would involve acting as an additional anchor (Fig. 3 A). In some systems, juxta-transmembrane palmitoylation allows tilting of transmembrane domains, effectively shortening the transmembrane domain to reduce hydrophobic mismatch (Nyholm et al., 2007), particularly at the thinner ER membrane (Abrami et al., 2008; Charollais and Van Der Goot, 2009; Baekkeskov and Kanaani, 2009), and confer conformational restraints on the peptide (Fig. 3 A). Such a mechanism has been proposed to control ER exit of the regulatory β4 subunits of BK channels. In this case, depalmitoylation of a cysteine residue juxtaposed to the second transmembrane domain of the β4 subunits may result in hydrophobic mismatch at the ER, reducing ER exit, and yield a conformation that is unfavorable for interaction with BK channel α subunits, thereby decreasing surface expression of BK channel α subunits (Chen et al., 2013).

Tools to analyze ion channel S-acylation

Before the seminal discovery of the mammalian enzymes that control S-acylation (Fukata et al., 2004) and current advances in proteomic techniques to assay S-acylation, progress in the field was relatively slow, largely because of the lack of pharmacological, proteomic, and genetic tools to investigate the functional role of S-acylation. It is perhaps instructive to consider that protein tyrosine phosphorylation was discovered the same year as S-acylation (Hunter, 2009). However, the subsequent rapid identification and cloning of tyrosine kinases provided a very extensive toolkit to investigate this pathway. Although the S-acylation toolkit remains limited, the last few years have seen rapid progress in our ability to interrogate S-acylation function and its control of ion channel physiology. Furthermore, S-acylation prediction algorithms, such as CSS-palm 4.0 (Ren et al., 2008), provide an in silico platform to inform experimental approaches for candidate targets.

Pharmacological tools.

The S-acylation pharmacological toolkit remains, unfortunately, empty, with limited specific agents with which to explore S-acylation function in vitro or in vivo. Although the palmitate analogue 2-bromopalmitate (2-BP) is widely used for cellular assays and to analyze ion channel regulation by S-acylation, caution must be taken in using this agent, even though it remains our best pharmacological inhibitor of zDHHCs (Resh, 2006; Davda et al., 2013; Zheng et al., 2013). Unfortunately, 2-BP is a nonselective inhibitor of lipid metabolism and many membrane-associated enzymes, and displays widespread promiscuity (e.g., Davda et al., 2013); does not show selectivity toward specific zDHHC proteins (Jennings et al., 2009); has many pleiotropic effects on cells at high concentrations, including cytotoxicity (Resh, 2006); and also inhibits acylthioesterases (Pedro et al., 2013). Other lipid inhibitors include cerulenin and tunicamycin. However, cerulenin affects many aspects of lipid metabolism, and tunicamycin inhibits N-linked glycosylation (Resh, 2006). Although some nonlipid inhibitors have been developed, these are not widely used (Ducker et al., 2006; Jennings et al., 2009), and there are currently no known activators of zDHHCs or compounds that inhibit specific zDHHCs. In the last few years, several inhibitors for the acylthioesterases LYPLA1 and LYPLA2 have been developed (Bachovchin et al., 2010; Dekker et al., 2010; Adibekian et al., 2012). However, several of these compounds, such as palmostatin B, are active against several members of the larger serine hydrolase family. Clearly, the development of novel S-acylation inhibitors and activators that display both specificity and zDHHC selectivity would represent a substantial advance for investigation of channel S-acylation.

Genetic tools.

To date, most studies have used overexpression of candidate zDHHCs in heterologous expression or native systems and analyzed increases in [3H]palmitate incorporation to define zDHHCs that may S-acylate specific ion channels (e.g. Rathenberg et al., 2004; Hayashi et al., 2005, 2009; Tian et al., 2010; Thomas et al., 2012). Although this is a powerful approach, caution is required to determine whether results obtained with overexpression in fact replicate endogenous regulation. For example, overexpression of some zDHHCs normally expressed in the cell type of interest can result in S-acylation of a cysteine residue that is not endogenously palmitoylated in BK channels (Tian et al., 2010). Point mutation of the cysteine of the catalytic DHHC domain abolishes the acyltransferase activity of zDHHCs and is thus an invaluable approach to confirming that the acyltransferase function of overexpressed zDHHC is required by itself. Increasingly, knockdown of endogenous zDHHCs using siRNA, and related approaches, is beginning to reveal the identity of zDHHCs that S-acylate native ion channel subunits. For example, knockdown of zDHHCs 5 or 8 reduces S-acylation of the accessory subunits PICK1 and Grip1, which control AMPA receptor trafficking (Thomas et al., 2012, 2013); and knockdown of zDHHC2 disrupts local nanoclusters of the PDZ domain protein PSD-95 in neuronal dendrites to control AMPA receptor membrane localization (Fukata et al., 2013). However, relatively few studies have taken a systematic knockdown approach to identify zDHHCs important for ion channel S-acylation. One such approach has, however, revealed that multiple, distinct zDHHCs mediate palmitoylation of the BK channel C terminus (zDHHCs 3, 5, 7, 9, and 17) and that a different subset of zDHHCs (22 and 23) mediate S-acylation of the intracellular S0-S1 loop in the same channel (Tian et al., 2010, 2012). Because some zDHHCs are themselves palmitoylated, the functional effect of overexpressing or knocking down individual zDHHCs on the localization and activity of other zDHHCs must also be carefully determined. For example, siRNA-mediated knockdown of zDHHC 5, 7, or 17 in HEK293 cells paradoxically results in an up-regulation of zDHHC23 mRNA expression (Tian et al., 2012). Furthermore, because many signaling and cytoskeletal elements are also controlled by S-acylation, direct effects on channel S-acylation by themselves must be evaluated in parallel (for example using site-directed cysteine mutants of the channel subunit). Fewer studies have used these approaches to examine the role of acylthioesterases, although overexpression of LYPLA1 and a splice variant of LYPLAL1, but not LYPLA2, deacylates the S0-S1 loop of the BK channel, promoting Golgi retention of the channels (Tian et al., 2012). Gene-trap and knockout mouse models for some zDHHCs (such as 5 and 17) are becoming available, although full phenotypic analysis and analysis of ion channel function in these models are largely lacking.

Proteomic and imaging tools. Lipid-centric (metabolic) labeling assays.

Metabolic labeling approaches are most suited to analysis of isolated cells, rather than tissues, but provide information on dynamic palmitoylation of proteins during the relatively short (∼4 h) labeling period as well as insight into the species of lipid bound to cysteine residues. The classical approach using radioactive palmitate (e.g., [3H]palmitate) remains a “gold standard” for validation, in particular for identification that palmitate is the bound lipid. However, metabolic labeling with [3H]palmitate generally requires immunoprecipitation and days to weeks of autoradiography or fluorography, particularly when analyzing low abundance membrane proteins such as ion channels. To overcome some of these issues, and also to provide a platform to allow cellular imaging of S-acylation, a variety of biorthogonal lipid probes have recently been developed (Hannoush and Arenas-Ramirez, 2009; Hannoush, 2012; Martin et al., 2012; for reviews see Charron et al., 2009a; Hannoush and Sun, 2010). These probes are modified fatty acids with reactive groups, such as an azide or alkyne group, allowing labeled proteins to be conjugated to biotin or fluorophores via the reactive group using Staudinger ligation or “click” chemistry. In particular, development of a family of ω-alkynyl fatty acid probes of different chain lengths (such as Alk-C16 and Alk-C18) have been exploited for proteomic profiling as well as single cell imaging (Gao and Hannoush, 2014) and have been used to identify candidate S-acylated channels in several mammalian cell lines (Charron et al., 2009b; Hannoush and Arenas-Ramirez, 2009; Martin and Cravatt, 2009; Yap et al., 2010; Yount et al., 2010; Martin et al., 2012). It is important to note that palmitic acid can also be incorporated into free N-terminal cysteines of proteins via an amide linkage (N-palmitoylation), addition of the monounsaturated palmitoleic acid via an oxyester linkage to a serine residue (O-palmitoylation), and oleic acid (oleoylation) as well as myristate via amide linkages on lysine residues (Stevenson et al., 1992; Linder and Deschenes, 2007; Hannoush and Sun, 2010; Schey et al., 2010). These modifications can be discriminated from S-acylation by their insensitivity to hydroxylamine cleavage (at neutral pH) compared with the S-acylation thioester linkage. Whether N- or O-linked palmitoylation or oleoylation controls ion channel function remains to be determined.

Cysteine centric (cysteine accessibility) assays: Acyl-biotin exchange (ABE) and resin-assisted capture (Acyl-RAC).

The metabolic labeling approach requires treating isolated cells with lipid conjugates and thus largely precludes analysis of native S-acylation in tissues. However, several related approaches have been developed that exploit the exposure of a reactive cysteine after hydroxylamine cleavage (at neutral pH) of the cysteine-acyl thioester linkage. The newly exposed cysteine thiol can then react with cysteine-reactive groups (such as biotin-BMCC or biotin-HPDP used in the ABE approach; Drisdel and Green, 2004; Drisdel et al., 2006; Draper and Smith, 2009; Wan et al., 2007) or thiopropyl sepharose (used in Acyl-RAC; Forrester et al., 2011) to allow purification of S-acylated proteins that can be identified by Western blot analysis or mass spectrometry. Acyl-RAC has been reported to improve detection of higher molecular weight S-acylated proteins and thus may prove valuable for ion channel analysis. These approaches have been exploited to determine the “palmitoylome” in several species and tissues (e.g., Wan et al., 2007, 2013; Kang et al., 2008; Martin and Cravatt, 2009; Yang et al., 2010; Yount et al., 2010; Merrick et al., 2011; Wilson et al., 2011; Jones et al., 2012; Ren et al., 2013). For example, analysis of rat brain homogenates identified both previously characterized as well as novel S-acylated ion channels (Wan et al., 2013), although it must be remembered that these approaches detect S-acylation and do not define S-palmitoylation per se. Cysteine accessibility approaches determine the net amount of preexisting S-acylated proteins; however, caution is required to eliminate false positives. In particular it is necessary to fully block all reactive cysteines before hydroxylamine cleavage; moreover, the identity of the endogenously bound lipid is of course not known.The lipid- and cysteine-centric approaches are thus complementary. In conjunction with site-directed mutagenesis of candidate S-acylated cysteine residues in ion channel subunits, these approaches have provided substantial insight into the role and regulation of ion channel S-acylation (Fukata et al., 2013). However, this approach does not directly confirm that the protein is S-acylated per se. Furthermore, in most ion channels, and in fact most S-acylated proteins, the identity of the native lipid bound to a specific S-acylated cysteine is not known. Although palmitate is considered to be the major lipid species involved in S-acylation, this has not been directly demonstrated in most cases, and other fatty acids, including arachidonic acid, oleate acid, and stearic acid, have also been reported to bind to cysteine via a thioester S-linkage (Linder and Deschenes, 2007; Hannoush and Sun, 2010). A major reason for this discrepancy is that mass spectrometry–based approaches to identify the native lipid specifically bound to S-acylated cysteines remain a significant challenge. This is particularly true for low abundance proteins such as mammalian ion channels, in contrast to the widespread application of mass spectrometry to directly identify native amino acids that are phosphorylated (Kordyukova et al., 2008, 2010; Sorek and Yalovsky, 2010; McClure et al., 2012; Ji et al., 2013). As such, direct biochemical demonstration of native cysteine S-acylation is lacking in most ion channels.

S-acylation and control of the ion channel lifecycle

Ion channel physiology is determined by both the number of channel proteins at the cognate membrane and by their activity and/or kinetics at the membrane. Evidence has begun to emerge that S-acylation of either pore-forming or regulatory subunits of ion channels controls all of these aspects of ion channel function. Although the focus of this review is S-acylation–dependent regulation of ion channel subunits itself, S-acylation also regulates the localization or activity of many adaptor, scaffolding, and cellular signaling proteins (e.g., G protein–coupled receptors [GPCRs], AKAP18, AKAP79/150, G proteins, etc.), as well as other aspects of cell biology that affect ion channel trafficking and the activity and regulation of macromolecular ion channel complexes (El-Husseini and Bredt, 2002; Linder and Deschenes, 2007; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Shipston, 2011; Resh, 2012).

Control of ion channel cell surface expression and spatial organization in membranes.

The control of ion channel trafficking, from synthesis in the ER through modification in the Golgi apparatus to subsequent delivery to the appropriate cellular membrane compartment, is a major mechanism whereby S-acylation modulates ion channel physiology. S-acylation may influence the number of ion channels resident in a membrane through regulation of distinct steps in the ion channel lifecycle (Fig. 2). Indeed S-acylation has been implicated in ion channel synthesis, as well as in channel trafficking to the membrane and subsequent internalization, recycling, and degradation. S-acylation controls the maturation and correct assembly of ion channels early in the biosynthetic pathway. For example, S-acylation regulates assembly of the ligand gated nicotinic acetylcholine receptor (nAChR) to ensure a functional binding site for acetylcholine (Alexander et al., 2010) as well as controlling its surface expression (Amici et al., 2012). S-acylation is also an important determinant of the maturation of both voltage-gated sodium (Nav1.2) and voltage-gated potassium channels (Kv1.5; Schmidt and Catterall, 1987; Zhang et al., 2007). S-acylation also contributes to the efficient trafficking of channels from the ER to Golgi and to post-Golgi transport. Three examples illustrate the importance and potential complexity of S-acylation in controlling ion channel trafficking:(1) S-acylation of a cysteine residue adjacent to a hydrophobic region (TM2) in a cytosolic loop of the GluA1 pore-forming subunit of AMPA receptors (Fig. 4 A) promotes retention of the channel in the Golgi (Hayashi et al., 2005). However, S-acylated Grip1b, a PDZ protein that binds to AMPA receptors, is targeted to mobile trafficking vesicles in neuronal dendrites and accelerates local recycling of AMPA receptors to the plasma membrane (Thomas et al., 2012). In contrast, S-acylation of another AMPA receptor interacting protein, PICK1, is proposed to stabilize AMPA receptor internalization (Thomas et al., 2013).(2) S-acylation of a cluster of cysteine residues juxtaposed to the transmembrane 4 domain (Cys I site) of the NMDA receptor subunit GluN2A (Fig. 4 B) increases surface expression of NMDA receptors by decreasing their constitutive internalization. In contrast S-acylation at C-terminal cysteine residues (Cys II site) decreases their surface expression by introducing a Golgi retention signal that decreases forward trafficking (Hayashi et al., 2009). Even though both sites affect surface expression, only S-acylation of the TM4 juxtaposed cysteine residues influences synaptic incorporation of NMDA receptors, which suggests that this site is an important determinant of the synaptic versus extrasynaptic localization of these ion channels (Mattison et al., 2012). Together, these data highlight the importance of S-acylation of two distinct sites within the same ion channel as well as that of components of the ion channel multimolecular complex as determinants of channel trafficking.(3) S-acylation of a cluster of cysteine residues in the intracellular S0-S1 loop of the pore-forming subunit (Figs. 4 C and and5)5) is required for efficient exit of BK channels from the ER and the trans-Golgi network. Deacylation at the Golgi apparatus appears to be an important regulatory step (Tian et al., 2012). BK channel surface abundance may also be controlled by S-acylation of regulatory β4 subunits. β4 subunit S-acylation on a cysteine residue juxtaposed to the second transmembrane domain is important for the ability of the β4 subunit itself to exit the ER. Importantly, assembly of β4 subunits with specific splice variants of pore-forming α subunits of the BK channel enhances surface expression of the channel, a mechanism that depends on S-acylation of the β4 subunit (Chen et al., 2013). Thus, in BK channels, S-acylation of the S0-S1 loop of the pore-forming subunit controls global BK channel surface expression, and β4 subunit S-acylation controls surface expression of specific pore-forming subunit splice variants. S-acylation of the Kchip 2 and Kchip 3 accessory subunits also controls surface expression of voltage-gated Kv4.3 channels (Takimoto et al., 2002).Moreover, S-acylation modulates the spatial organization of ion channels within membranes. Perhaps the most striking example involves aquaporin 4 (AQP4), where S-acylation of two N-terminal cysteine residues in an N-terminal splice variant (AQP4M1) inhibits assembly of AQP4 into large orthogonal arrays (Suzuki et al., 2008; Crane and Verkman, 2009), perhaps by disrupting interactions within the AQP4 tetramer. S-acylation can affect the distribution of the many membrane-associated proteins between cholesterol-rich microdomains (lipid rafts) and the rest of the membrane. Such clustering has also been reported for various transmembrane proteins, including the P2x purinoceptor 7 (P2X7) receptor, in which S-acylation of the C terminus promotes clustering into lipid rafts (Gonnord et al., 2009). A similar mechanism may underlie synaptic clustering of GABAA receptors mediated by S-acylation of an intracellular loop of the y2 subunit (Rathenberg et al., 2004). In these examples, S-acylation of the channel itself affects membrane partitioning and organization. However, recent evidence in neurons suggests that establishment of “nano” domains of ion channel complexes in postsynaptic membranes may also be established by local clustering of the cognate acyltransferase itself. For example, clustering of zDHHC2 in the postsynaptic membranes of individual dendritic spines provides a mechanism for local control of S-acylation cycles of the PDZ protein adapter, PSD-95, and thereby for controlling its association with the plasma membrane. PSD-95, in turn, can assemble with various ion channels, including NMDA receptors, and can thus dynamically regulate the localization and clustering of ion channel complexes (Fukata et al., 2013). Indeed, an increasing number of other ion channel scaffolding proteins such as Grip1 (Thomas et al., 2012), PICK1 (Thomas et al., 2013), S-delphilin (Matsuda et al., 2006), and Ankyrin G (He et al., 2012) that influence ion channel trafficking, clustering, and localization are now known to be S-acylated.Relatively few studies have identified effects of S-acylation on the intrinsic gating kinetics or pharmacology of ion channels at the plasma membrane. However, a glycine-to-cysteine mutant (G1079C) in the intracellular loop between domains II and III enhances the sensitivity of the voltage-gated Na channel Nav1.2a to the toxins PaurTx3 and ProTx-II, an effect blocked by inhibition of S-acylation. These toxins control channel activation through the voltage sensor in domain III. In addition, deacylation of another (wild-type) cysteine residue (C1182) in the II–III loop produces a hyperpolarizing shift in both activation and steady-state inactivation as well as slowing the recovery from fast inactivation and increasing sensitivity to PaurTx3 (Bosmans et al., 2011). Effects of S-acylation on gating kinetics have also been reported in other channels. For example, in the voltage-sensitive potassium channel Kv1.1, S-acylation of the intracellular linker between transmembrane domains 2 and 3 increases the intrinsic voltage sensitivity of the channel (Gubitosi-Klug et al., 2005). S-acylation of the β and γ subunits of epithelial sodium channels (ENaC) also affects channel gating (Mueller et al., 2010; Mukherjee et al., 2014), and the S-acylated regulatory β2a subunit of N-type calcium channels controls voltage-dependent inactivation (Qin et al., 1998; Hurley et al., 2000).S-acylation is also an important determinant of retrieving ion channels from the plasma membrane for recycling or degradation. S-acylation of a single cysteine residue juxtaposed to the transmembrane TM4 domain of GluA1 and GluA2 subunits of AMPA receptors controls agonist-induced ion channel internalization. These residues are distinct from those controlling Golgi retention of AMPA receptors (Fig. 4 A), which emphasizes the finding that the location and context of the S-acylated cysteines, even in the same protein, is central for their effects on physiological function (Hayashi et al., 2005; Lin et al., 2009; Yang et al., 2009). The stability of many proteins is also regulated by S-acylation; S-acylation of a single cysteine residue in Kv1.5 promotes both its internalization and its degradation (Zhang et al., 2007; Jindal et al., 2008). Thus, in different ion channels, S-acylation can have opposite effects on insertion, membrane stability, and retrieval.

S-acylation and posttranslational cross-talk control channel trafficking and activity.

An emerging concept is that S-acylation is an important determinant of ion channel regulation by other PTMs. Indeed, nearly 20 years ago it was reported that PKC-dependent phosphorylation of the GluK2 (GluR6) subunit of Kainate receptors was attenuated in channels S-acylated at cysteine residues near the PKC consensus site (Pickering et al., 1995). S-acylation of GluA1 subunits of AMPA receptors also blocks PKC phosphorylation of GluA1 and subsequently prevents its binding to the cytoskeletal adapter protein 4.1N, ultimately disrupting AMPA receptor insertion into the plasma membrane (Lin et al., 2009). Intriguingly, PKC phosphorylation and S-acylation have the opposite effect on 4.1N-mediated regulation of Kainate receptor (GluK2 subunit) membrane insertion: in this, case S-acylation promotes 4.1N interaction with Kainate receptors and thereby receptor insertion, whereas PKC phosphorylation disrupts 4.1N interaction, promoting receptor internalization (Copits and Swanson, 2013). Disruption of phosphorylation by S-acylation of residues near consensus phosphorylation sites likely results from steric hindrance, as proposed for S-acylation–dependent regulation of β2 adrenergic receptor phosphorylation (Mouillac et al., 1992; Moffett et al., 1993).S-acylation has also been reported to promote ion channel phosphorylation. For example, site-directed mutation of a cluster of palmitoylated cysteine residues in the GluN2A subunit of NMDA receptors abrogates Fyn-dependent tyrosine phosphorylation at a site between TM4 and the palmitoylated cysteines (Hayashi et al., 2009). Therefore, S-acylation of GluN2A promotes tyrosine phosphorylation, resulting in reduced internalization of the NMDA receptor (Hayashi et al., 2009). Furthermore, S-acylation of BK channels can act as a gate to switch channel regulation to different AGC family kinase signaling pathways, emphasizing the complex interactions that can occur between signaling pathways (Tian et al., 2008; Zhou et al., 2012; Fig. 5). S-acylation of an alternatively spliced insert (STREX) in the large cytosolic domain of the pore-forming subunit of BK channels promotes association of the STREX domain with the plasma membrane. S-acylation of the STREX insert is essential for the functional inhibition of STREX BK channels by PKA-mediated phosphorylation of a serine residue immediately upstream of the S-acylated cysteines. PKA phosphorylation dissociates the STREX domain from the plasma membrane (Tian et al., 2008), preventing STREX domain S-acylation (Jeffries et al., 2012) and leading to channel inhibition. However, deacylation of the STREX domain exposes a PKC consensus phosphorylation site downstream of the STREX domain, allowing PKC to inhibit STREX BK channels (Zhou et al., 2012). Thus, S-acylation acts as a reversible switch to specify regulation by AGC family kinases through control of the membrane association of a cytosolic domain of the channel: S-acylated STREX BK channels are inhibited by PKA but insensitive to PKC, whereas deacylated channels are inhibited by PKC but not PKA (Fig. 5). The reciprocal control of membrane association of a protein domain by S-acylation and protein phosphorylation likely represents a common mechanism in other signaling proteins as revealed for phosphodiesterase 10A (Charych et al., 2010).Cysteine residues are targets for several other modifications that regulate various ion channels, including nitrosylation, sulphydration, REDOX regulation, and formation of disulphide bonds (Sen and Snyder, 2010). Evidence is beginning to emerge that S-acylation may mutually compete with these mechanisms, providing a dynamic network to control cysteine reactivity. For example, the ion channel scaffolding PDZ domain protein PSD-95 is S-acylated at two N-terminal cysteine residues (C3 and C5) that are required for membrane targeting and clustering of PSD-95 (El-Husseini et al., 2002). nNOS also interacts with PSD-95, and stimulation of nitric oxide production results in nitrosylation of these cysteines, preventing their S-acylation and thereby decreasing PSD-95 clusters at postsynaptic sites (Ho et al., 2011). A recent remarkable example of the potential for such cross-talk in ion channel subunits is the identification of the S-acylation of 18 different cysteine residues in the large cytosolic N terminus of RyR1 in skeletal muscle. Of these 18 S-acylated cysteines, six have previously been identified as targets for S-oxidation, and a further cysteine residue was also subject to S-nitrosylation (Chaube et al., 2014) Although the functional relevance of this potential cross-talk in RyR1 has yet to be defined, interaction between oxidation and S-acylation of the same cysteine residue is physiologically relevant in other proteins. For example, oxidation of the signaling protein HRas at two cysteine residues C181/184 prevents S-acylation of these residues, resulting in a loss of plasma membrane localization of this peripheral membrane signaling protein (Burgoyne et al., 2012). Intriguingly, a conserved cysteine residue in nAChR α3 subunits, which has been shown to be S-acylated (C273) in the nAChR α4 subunit, has been implicated in use-dependent inactivation of nAChRs by reactive oxygen species (Amici et al., 2012). Determining whether these mutually competitive cysteine modifications represent an important mechanism for regulation of a range of ion channels is an exciting challenge for the future.S-acylation is also an important determinant of ion channel regulation by heterotrimeric G proteins. This can involve S-acylation of either G protein targets or of regulators of G proteins. In an example of the former, the palmitoylated N terminus of the regulatory β2a subunit splice variant acts as a steric inhibitor of an arachidonic acid binding domain to stimulate N-type calcium channels (Chien et al., 1996; Heneghan et al., 2009; Mitra-Ganguli et al., 2009). When the regulatory β subunits are not S-acylated, however, Gq-mediated signaling, via arachidonic acid, inhibits calcium channel activity. Closure of G protein regulated inward rectifying potassium (GIRK) channels in neurons after Gi/o deactivation provides an example of the latter (Jia et al., 2014). Signaling by members of the Gi/o family of the Gα subunit of heterotrimeric G proteins is terminated by members of the regulator of G protein signaling 7 (R7 RGS) family of GTPase-activating proteins, which accelerate GTP hydrolysis to speed Gi/o deactivation. Membrane localization of regulator of G protein signaling 7 (R7-RGS) is required for its regulation of Gi/o, and this is determined by interaction with an S-acylated R7 binding protein (R7-BP) that acts as an allosteric activator. Thus, the R7-RGS complex, recruited to the plasma membrane by S-acylated R7-BP, promotes Gi/o deactivation to facilitate GIRK channel closure. Conversely, deacylation of R7-BP removes the R7-GS complex from the plasma membrane, slowing Gi/o deactivation and consequent channel closure (Jia et al., 2014). Clearly, as S-acylation can also control an array of GPCRs, enzymes, and signaling and adapter proteins that indirectly control ion channel function (El-Husseini and Bredt, 2002; Linder and Deschenes, 2007; Fukata and Fukata, 2010; Greaves and Chamberlain, 2011; Shipston, 2011; Resh, 2012), understanding how S-acylation dynamically controls other components of ion channel multimolecular signaling complexes will be an essential future goal.

Summary and perspectives

With an ever-expanding “catalog” of S-acylated ion channel pore-forming and regulatory subunits (∼50 to date), together with an array of S-acylated scaffolding and signaling proteins, the importance and ubiquity of this reversible covalent lipid modification in controlling the lifecycle and physiological function and regulation of ion channels is unquestionable. This has been paralleled by a major resurgence in the wider S-acylation field, a consequence in large part of the discovery of S-acylating and deacylating enzymes together with a growing arsenal of genetic, proteomic, imaging, and pharmacological tools to assay and interrogate S-acylation function.As for most other posttranslational modifications of ion channels, including phosphorylation, major future goals for the field include:(1) Understanding mechanistically how covalent addition of a fatty acid can control such a diverse array of ion channel protein properties and functions, and how this is spatiotemporally regulated.(2) Elucidating the physiological relevance of this posttranslational modification from the level of single ion channels to the functional role of the channel in the whole organism in health and disease.Elucidation of these issues has fundamental implications far beyond ion channel physiology.To address these goals several major challenges and questions must be addressed, including:(1) It is largely assumed that S-acylation of transmembrane proteins results in an additional “membrane anchor” to target domains to the membrane interface. However, understanding the mechanisms, forces, and impact of S-acylation on the orientation of transmembrane helices and the architecture and structure of disordered domains in cytosolic loops and linkers, while remaining a considerable technical challenge, should provide major insight into mechanisms controlling channel trafficking, activity, and regulation.(2) Although S-acylation is widely accepted to be reversible, its spatiotemporal regulation of most ion channels is unknown. Mechanistic insight into zDHHC and acylthioesterase substrate specificity, native subcellular localization, and assembly with ion channel signaling complexes will allow us to dissect and understand how S-acylation of ion channels is controlled. Importantly, this should allow us to take both “channel-centric” (e.g., site-directed mutagenesis of S-acylated cysteines) as well as “S-acylation centric” (e.g., knockout of specific zDHHC activity) approaches to understand how multisite S-acylation on the same ion channel subunit can control distinct functions as well as physiological regulation of trafficking and function at the plasma membrane.(3) The functional role of S-acylation cannot be viewed in isolation from other posttranslational modifications. The cross-talk between S-acylation and adjacent phosphorylation sites as well as other cysteine modifications highlights the importance of understanding the interactions between signaling pathways. Insight into the rules, mechanisms, and cross-talk of S-acylation with these modifications has broad implications for cellular signaling.(4) Although it is clear that disruption of S-acylation homeostasis itself has substantial effects on normal physiology, and we are beginning to understand some of the cellular functions of ion channel S-acylation, we know very little about the functional impact of disrupted ion channel S-acylation at the systems and organismal level. Understanding how this may be dynamically regulated during a lifespan is critical to understanding the role of S-acylation in health and disease.To address these issues, development of improved tools to assay and investigate S-acylation from the single protein to organism is required. For example, tools to allow the real-time analysis of S-acylation status of ion channels in cells and tissues will provide fundamental insights into its dynamics and role in ion channel trafficking and membrane localization. Improved proteomic tools will allow direct assay of fatty acids bound to cysteine residues via thioester linkages. Development of new tools and models are essential if we are to understand the physiological relevance of ionic channel S-acylation at the systems level. These include: specific inhibitors of zDHHCs and thioesterases, conditional knockouts to spatiotemporally control zDHHC expression, and transgenics expressing catalytically inactive zDHHCs and models expressing S-acylation–null ion channel subunits. Furthermore, our understanding of how S-acylation may be dynamically controlled during normal ageing in response to homeostatic challenge and disruption in disease states remains rudimentary. Whether we will start to uncover channel “S-acylationopathies” resulting from dysregulation of ion channel S-acylation, analogous to channel phosphorylopathies, remains to be explored. Addressing these issues, together with development of new tools, will provide a paradigm shift in our understanding of both ion channel and S-acylation physiology, and promises to reveal novel therapeutic strategies for a diverse array of disorders.  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