Friday, September 16, 2022
HomeBiologyMolecular structure of the C. elegans centriole

Molecular structure of the C. elegans centriole


Summary

Uncovering organizing ideas of organelle meeting is a basic pursuit within the life sciences. Caenorhabditis elegans was key in figuring out evolutionary conserved elements governing meeting of the centriole organelle. Nonetheless, localizing these elements with excessive precision has been hampered by the minute dimension of the worm centriole, thus impeding understanding of underlying meeting mechanisms. Right here, we used Ultrastructure Growth coupled with STimulated Emission Depletion (U-Ex-STED) microscopy, in addition to electron microscopy (EM) and electron tomography (ET), to decipher the molecular structure of the worm centriole. Attaining an efficient lateral decision of roughly 14 nm, we localize centriolar and PeriCentriolar Materials (PCM) elements in a complete method with utmost spatial precision. We discovered that every one 12 elements analysed exhibit a ring-like distribution with distinct diameters and sometimes with a 9-fold radial symmetry. Furthermore, we uncovered that the procentriole assembles at a location on the centriole margin the place SPD-2 and ZYG-1 additionally accumulate. Furthermore, SAS-6 and SAS-5 have been discovered to be current within the nascent procentriole, with SAS-4 and microtubules recruited thereafter. We registered U-Ex-STED and EM information utilizing the radial array of microtubules, thus permitting us to map every centriolar and PCM protein to a particular ultrastructural compartment. Importantly, we found that SAS-6 and SAS-4 exhibit a radial symmetry that’s offset relative to microtubules, resulting in a chiral centriole ensemble. Moreover, we established that the centriole is surrounded by a area from which ribosomes are excluded and to which SAS-7 localizes. Total, our work uncovers the molecular structure of the C. elegans centriole in unprecedented element and establishes a complete framework for understanding mechanisms of organelle biogenesis and performance.

Introduction

Centrioles are membrane-less organelles that have been current within the final frequent ancestor of eukaryotes (reviewed in [1]). In cells with flagella or cilia, centrioles act as basal our bodies that template the formation of those constructions. Furthermore, in animal cells, centrioles kind the core of the centrosome, which organizes microtubules and is thereby vital for basic mobile processes, together with polarity and division (reviewed in [2]). In most organisms, centrioles are cylindrical organelles roughly 500 nm excessive and roughly 250 nm broad, with a 9-fold radially symmetric distribution of microtubules (reviewed in [3,4]). These centriolar microtubules are organized in triplets within the proximal area of the organelle and in doublets extra distally. Triplet and doublet microtubules are twisted in a clockwise route with respect to the microtubules when seen from the distal finish of the centriole, ensuing within the attribute chiral geometry of the organelle. This 9-fold radially symmetric structure can also be imparted onto the ciliary and flagellar axoneme that stem from centriolar microtubules and is likely to be evolutionarily conserved as a result of it offers an optimum geometry for axonemal motility. Regardless of essential progress in recent times, the detailed molecular structure of the centriole, together with the basis of its attribute chirality, stays incompletely understood.

There are variations within the architectural options of centrioles in some programs, that are normally correlated with the absence or discount of ciliary and flagellar motility (reviewed in [5]). As an illustration, within the nematode Caenorhabditis elegans, motile cilia and flagella are absent, and the sperm strikes in an amoeboid trend. Maybe within the absence of evolutionary stress for ciliary and flagellar motility, centrioles are smaller (roughly 175 nm excessive and roughly 120 nm broad) within the embryo [68] and comprise a radial association of 9 microtubule singlets as a substitute of the same old triplets and doublets [9]. Electron microscopy (EM) of centrioles within the C. elegans embryo revealed ultrastructural compartments in addition to microtubules, together with 9 peripheral paddlewheels, in addition to the central tube and, extra centrally nonetheless, the interior tube [68]. EM evaluation of embryonic centrioles additionally led to the notion that every paddlewheel is offset with respect to its accompanying microtubule, with a clockwise twist when seen from the distal finish, leading to a chiral ensemble [8]. Whether or not chirality of the C. elegans centriole is obvious extra centrally within the organelle, the place the meeting course of is believed to provoke, isn’t identified.

As in different programs, beginning roughly on the onset of S section, the two resident centrioles in C. elegans every seed the meeting of a procentriole of their neighborhood, such that 4 centriolar models are current throughout mitosis, 2 per spindle pole. Complete genetic and useful genomic screens carried out in C. elegans led to the invention of 6 elements important for procentriole formation (reviewed in [1012]). Molecular epistasis experiments uncovered the order wherein proteins important for procentriole formation are recruited to the worm organelle [7,13]. These experiments established that SAS-7 and SPD-2 (Cep192 in people) are first recruited to the resident centriole. Thereafter, the kinase ZYG-1 (Plk4 in people) directs the interacting coiled-coil proteins SAS-6 (HsSAS-6 in people) and SAS-5 (STIL in people) to the procentriole meeting web site. That is adopted by SAS-4 (CPAP in human) recruitment to the procentriole, a protein thought to allow the addition of microtubules to the SAS-6/SAS-5 scaffold. Relations of SPD-2, ZYG-1, SAS-6, SAS-5, and SAS-4 in different programs are recruited in the same sequence and exert analogous capabilities in procentriole formation (reviewed in [1012]).

SAS-6 is the primary constructing block of a scaffold known as the cartwheel, which is believed to contribute to imparting the 9-fold radial symmetry of the organelle [14,15]. Whereas SAS-6 proteins in different programs self-assemble into ring-containing polymers that stack to kind the cartwheel, structural and biophysical proof obtained with the C. elegans protein has led to the suggestion that SAS-6 varieties a steep spiral [16]. Nonetheless, whether or not that is the case in vivo has not been addressed.

HYLS-1 and SAS-1 are 2 extra C. elegans centriolar proteins which can be dispensable for procentriole meeting. Nonetheless, HYLS-1 is required for producing nonmotile cilia [17], whereas SAS-1 is vital for sustaining the integrity of the organelle as soon as shaped [18]. As well as, the Polo-like kinase PLK-1 is current at centrioles within the early worm embryo [19]. As in different programs, C. elegans centrioles recruit the PeriCentriolar Materials (PCM), thus forming the centrosome, which acts as a microtubule organizing heart (reviewed in [20]). Meeting of the C. elegans PCM core, which has been outlined because the set of PCM proteins which can be additionally current in interphase [21], depends on the interacting proteins SPD-2 [22,23] and SPD-5 [24], in addition to on SAS-7 [8] and PCDM-1 [21]. Moreover, the γ-tubulin protein TBG-1 [25,26], along with the γ-tubulin interacting proteins GIP-1 and GIP-2 [26], in addition to the γ-tubulin associate MZT-1 [27] are current within the worm PCM core. Further proteins, together with PLK-1 and AIR-1 [28,29], in addition to TAC-1 and ZYG-9 [30,31], are recruited to this PCM core when the centriole matures in mitosis within the embryo, resulting in elevated microtubule nucleation. Regardless of the most likely near-comprehensive checklist of element components of the centriole and the PCM core in C. elegans, the very small dimensions of the worm organelle have so far prevented localizing with precision the place every element resides, thus limiting understanding of how they operate.

The molecular structure of the centrioles has been investigated utilizing 3D-Structured Illumination Microscopy (SIM) or STED super-resolution microscopy in different programs the place the organelle is bigger than in C. elegans, together with human cells and Drosophila [3234]. Furthermore, ultrastructure growth (U-Ex) microscopy has been utilized to analyze the molecular structure of centrioles from human cells [35,36]. On this technique, the pattern is embedded in a gel that’s then expanded isotropically a number of fold, thus likewise increasing the efficient decision [37]. SIM, STED, and U-Ex have enabled putting in a extra refined method a subset of elements within the centriole map in these programs. Nonetheless, the decision achieved with these approaches can be possible inadequate to resolve the molecular structure of the minute worm centriole.

Right here, we got down to map in a complete method and with utmost precision the distribution of centriolar in addition to PCM core element within the gonad of C. elegans. Contemplating the very small dimension of the worm centriole, we mixed U-Ex and STED, reaching an efficient lateral decision of roughly 14 nm. Utilizing primarily endogenously tagged elements and validated antibodies, we might thus decide with beautiful precision the localization of 12 centriolar and PCM core proteins. Of specific curiosity, this revealed that SAS-6 and SAS-4 exhibit an angular offset with respect to the microtubules, leading to a chiral association within the organelle heart. Furthermore, we acquired a big corresponding EM information set, which we overlaid with the U-Ex-STED pictures to map every centriolar protein to a particular ultrastructural compartment of the organelle. Total, we uncovered the molecular structure of the C. elegans centriole and supply an unprecedented framework for a mechanistic dissection of centriole meeting and performance.

Outcomes

Combining nuclei spreading and U-Ex microscopy for improved decision of centrioles

We got down to analyze the molecular structure of the C. elegans centriole with utmost spatial decision, utilizing the grownup hermaphrodite gonad as an experimental system (Fig 1A). The distal a part of the syncytial gonad (the “mitotic zone” from right here on) constitutes a stem cell pool the place nuclei bear cell cycles characterised by brief G1 and M phases, with merely roughly 2% of nuclei being in certainly one of these 2 phases mixed [38]. As soon as nuclei have traveled far sufficient from the distal finish of the gonad, they bear premeiotic S section and enter meiotic prophase I, a protracted G2 section throughout which meiotic recombination happens.

thumbnail

Fig 1. Combining gonad nuclei spreading and U-Ex microscopy to research worm gonad centrioles.

(A) Widefield imaging of ethanol-fixed worm expressing GFP::SAS-7. One layer of nuclei of the gonad is max depth Z-projected (on this case, a top of 6.25 μm). White, gray, and purple daring dashed strains point out development by the gonad from the mitotic zone to early after which late meiotic prophase; different white dashed line outlines the gonad. Gray field is magnified in (B). Yellow dashed areas mark 3 oocytes, purple dashed area the spermatheca. Be aware that centrioles are eradicated in oocytes, previous to fertilization. (B) (Left) Magnification of gray field area from (A). (Center) Schematic illustration of a single nucleus proven within the left and proper panels. (Proper) Early prophase area of a diffusion gonad from a worm expressing GFP::SAS-7. Be aware that unfold nuclei are flattened and thus occupy a bigger space in comparison with not unfold nuclei. Be aware additionally that at this stage, centrioles don’t act as microtubule organizing facilities [55,56]. (C) Widefield imaging of centrioles within the early prophase area of the gonad from worms expressing GFP::SAS-7 earlier than (left) and after (proper) gel growth. Gray mesh within the background represents the gel matrix.


https://doi.org/10.1371/journal.pbio.3001784.g001

The gonad may be simply extruded from the animal and incorporates tons of of nuclei, that are virtually all in S or G2 phases of the cell cycle. Since procentriole formation begins in early S section, most gonad nuclei harbor 2 pairs of centriole/procentriole, that are in shut proximity to 1 one other and can’t be resolved by immunofluorescence (IF) in widefield microscopy, normally showing as a substitute as a single focus (Fig 1B, left). We took 2 steps to enhance the spatial decision for our evaluation. First, nuclei from extracted gonads have been adhered as a single layer to a coverslip utilizing delicate chromatin spreading [39], leading to superior detection by IF for the reason that specimen is nearer to the coverslip. Furthermore, the pool of cytoplasmic proteins, which might in any other case contribute to poor signal-to-noise ratio of the centriolar sign, is essentially washed out on this method (Fig 1B, proper). Second, we tailored beforehand validated ultrastructure gel growth strategies (U-Ex) [35,37], reaching roughly 5-fold isotropic growth of the specimens (Fig 1C, Supplies and strategies). Mixture of spreading with U-ExM enabled us to tell apart centriole and procentriole with widefield microscopy (S1A Fig), in addition to to localize elements to distinct areas inside the C. elegans centriole (Fig 1C, proper).

Procentriole meeting: Onset and maturation

We investigated the distribution of 12 centriolar and PCM core elements. As detailed in Desk 1 and the Supplies and strategies part, excluding mCherry::HYLS-1, we visualized every protein as an endogenously N-terminally [N] or C-terminally [C] tagged element, a tagged model expressed underneath the endogenous promoter within the absence of the endogenous element and/or beforehand validated antibodies towards the endogenous protein. We discovered that solely 3 of the 12 elements, SAS-6, SAS-5, and SAS-4, localize to each centriole and procentriole throughout S and G2. Utilizing sign depth in 3D-SIM pictures as a proxy for protein quantity, we discovered that there’s an undistinguishable quantity within the centriole and the procentriole for each SAS-5 and SAS-6 (Fig 2A). In distinction, the quantity of SAS-4 within the procentriole is on common roughly 10 occasions decrease than it’s within the centriole and in addition reveals massive variability (Fig 2A).

thumbnail

Fig 2. Composition and maturation of the procentriole.

(A) 3D-SIM sum depth Z-projected pictures of expanded centrioles from early meiotic prophase stained for the indicated proteins. White arrowheads level to centrioles, orange arrowheads to procentrioles. The presence of a ring-like distribution of RFP::SPD-2 (magenta) round one of many 2 inexperienced foci (SAS-6, SAS-5, or SAS-4) served to determine the centriole. The numbers under the photographs signify the ratio between the fluorescence of the indicated element within the centriole versus the procentriole (N = SAS-6: 8, SAS-5: 17, SAS-4: 18; ± signifies SD). Right here and in all different figures, scale bars inside a collection signify the identical size (e.g., 200 nm on this case). (B, C) (Left) U-Ex-STED of centrioles from early meiotic prophase revealing RFP::SPD-2 and SAS-5 distribution. RFP::SPD-2 indicators are displayed with the LUT “Hearth” (low intensities in blue, excessive intensities in magenta and purple). (Proper) Corresponding sign depth profiles alongside the dashed line depicted within the picture (10 pixels broad). The inexperienced bins within the pictures and the graphs point out RFP::SPD-2 situated underneath the procentriole recognized by SAS-5. Within the majority of circumstances, the RFP::SPD-2 sign is wider and brighter under the procentriole than anyplace else within the centriole (B, 22/33), whereas such enrichment couldn’t be detected within the the rest circumstances (C, 11/33). (D) U-Ex-STED of nuclei in both S-phase (left) or mitosis (proper) from the mitotic zone of the gonad. White arrowheads level to centrioles absolutely adorned with SAS-4 (mature centrioles). SAS-4 sign depth between the pair of centrioles current on every pole of the mitotic spindle varies by merely 14 ± 9% SD, N = 12 pairs. (E) Proportion of centrioles (recognized by SAS-6 surrounded by SPD-2) with or and not using a neighboring immature procentriole (recognized by SAS-6 not surrounded by SPD-2) in the course of the indicated levels. Nuclei in mitosis have been recognized by DNA staining as having probably the most condensed chromatin within the miotic area of the gonad; N = Mitotic zone (G1), S, G2-phase: 37, Mitosis (mitotic zone): 7, early meiotic prophase: 46. Knowledge underlying the graphs proven within the determine may be present in S1 Knowledge. SD, customary deviation; SIM, Structured Illumination Microscopy; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.


https://doi.org/10.1371/journal.pbio.3001784.g002

Along with the invariable presence of SAS-6 and SAS-5 on the procentriole, we discovered utilizing U-Ex-STED that ZYG-1 accumulates on the base of the procentriole in S and G2 within the mitotic area, in addition to throughout early meiotic prophase (S1B Fig). In distinction to SAS-6 and SAS-5, ZYG-1 ranges within the centrioles situated within the mitotic zone exhibit excessive variability. We speculate that ranges of ZYG-1 are larger in S section and decrease in G2 within the mitotic area as a result of low ranges have been additionally noticed in the course of the extended G2 of meiotic prophase (S1B Fig). Furthermore, we discovered SPD-2 to be current in a hoop across the centriole, abutting the bottom of the procentriole (Fig 2B). Curiously, SPD-2 radial distribution isn’t uniform however usually enriched on the web site of procentriole formation as evidenced by line scan evaluation (Fig 2B and 2C, 22/33 circumstances). We speculate that such an enrichment might replicate ZYG-1-mediated modification of SPD-2 to function a platform for procentriole formation or else native improve of SPD-2 on account of procentriole formation.

Earlier evaluation within the one-cell stage embryo established that the procentriole acquires SAS-4 and microtubules after SAS-6/SAS-5 recruitment [7]. Utilizing U-Ex-STED, we discovered within the gonad that the procentriole likewise harbors little SAS-4 initially and that extra protein is recruited at prometaphase, leading to comparable ranges of SAS-4 within the centriole and the procentriole by then (Fig 2D). This maturation coincides with the loading of microtubules onto the procentriole (S1C Fig). As anticipated from these observations, roughly 90% of centrioles harbor an immature procentriole throughout S and G2 phases within the mitotic area, whereas solely centrioles with out an accompanying procentriole are noticed by the point of mitosis, when the procentriole disengages and matures right into a centriole (Fig 2E). Moreover, in the course of the extended G2 of meiotic prophase that follows, all centrioles are once more accompanied by an immature procentriole (Fig 2E). These observations taken collectively point out that centriole formation within the gonad is characterised by 2 steps: an preliminary fast formation of a procentriole harboring SAS-6 and SAS-5, adopted briefly earlier than M section by the recruitment of different elements, together with SAS-4 and microtubules. Curiously, this coincides with the time throughout which centrioles recruit PCM and begin to set up the spindle (see under and reviewed in [20]), probably suggestive of a useful hyperlink between procentriole maturation and PCM growth.

U-Ex-STED reveals consecutive ring-like distribution of C. elegans centriolar proteins

We proceeded to comprehensively uncover the exact distribution of centriolar and PCM core proteins utilizing U-Ex-STED. We used prime views of centrioles to find out the radial distribution of those elements (Fig 3A). Remarkably, aside from ZYG-1 (see S1B Fig), such prime views revealed that every one elements exhibit a ring-like distribution, with distinct diameters. To research the place of every element with respect to the others, we decided the diameter of every ring relative to that of α-tubulin, which was used as an invariant reference on this evaluation (Fig 3B). To confirm the validity of this method, we costained α-tubulin with 2 completely different antibodies, discovering that the two indicators colocalize and that the corresponding rings therefore exhibit the identical diameter (with out correction for the growth issue: C-terminus 458 ± 38 nm, N-terminus 455 ± 35 nm, p = 0.81, N = 19; Fig 3A). Moreover, an antibody raised towards the center portion of SAS-4 likewise had the identical perimeter as α-tubulin, in step with the truth that the SAS-4-relative CPAP is a microtubule binding protein (Fig 3A and 3C, #9) [4042]. Thus, α-tubulin and SAS-4 can be utilized interchangeably as invariant references on this evaluation.

thumbnail

Fig 3. Relative place of centriolar elements inside the centriole.

(A) U-Ex-STED of centrioles from early meiotic prophase stained for the indicated proteins. Every element (inexperienced) was imaged along with both α-tubulin (visualized with an antibody recognizing the C-terminus of the protein) or SAS-4 (visualized with an antibody raised towards amino acid 350–517 of the protein) (each magenta). (B) Examples of fitted rings on fluorescent sign used to calculate the diameter of every element relative to α-tubulin or SAS-4 requirements. In every picture, the diameter of the centriolar element (in these circumstances RFP::SPD-2 (prime) and SPD-5 (center)) and that of the α-tubulin sign have been measured alongside the dashed strains. The perimeter of the centriolar element was then divided by that of the α-tubulin sign. To acquire the theoretical diameter of the element earlier than growth, this worth was normalized by the diameter of microtubules in EM pictures of centrioles (see Fig 5). (C) Calculated diameter of every centriolar element as decided in (B), organized from the smallest to the most important. Magenta field highlights α-tubulin (#8) and SAS-4 (#9). Numbers within the graph point out the id of the element. Colours point out whether or not the diameter is considerably completely different from 0 (purple), 1 (blue), or 2 (inexperienced) neighboring values (Scholar two-tailed t check, significance p < 0.005). The center strains of the boxplots correspond to the median, the cross represents the imply, the field contains 50% of values (IQR), and the whiskers present the vary of values inside 1.5*IQR. N = Flag::SAS-6: 21, SAS-5: 25, SAS-6: 22, SAS-1::FLAG: 10, SAS-4::GFP: 21, SAS-6::GFP: 15, FLAG::SAS-1 15, α-tubulin (N-ter): 19, SAS-4: 22, mCherrry::HYLS-1: 20, PCDM-1::GFP: 24, SPD-5: 20, GFP::PCDM-1: 21, SPD-2::GFP: 25, RFP::SPD-2: 20, GFP::SAS-7: 15, GFP::MZT-1: 20 and TBG-1: 20. Knowledge underlying the graphs proven within the determine may be present in S1 Knowledge. EM, electron microscopy; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.


https://doi.org/10.1371/journal.pbio.3001784.g003

To estimate the ring diameter of every element in nonexpanded samples, we decided the diameter of the ring shaped by the 9 microtubules in a novel EM information set of early meiotic prophase centrioles to be 87.9 ± 5.7 nm (N = 44; see under) and in contrast this worth to the α-tubulin sign diameter decided with U-Ex-STED (Fig 3B). Furthermore, we discovered that the α-tubulin diameter decided with U-Ex-STED following correction of the growth issue (5.2) is much like that measured for microtubules by EM (88 ± 8 nm; N = 38). This standardization technique enabled us to estimate the precise diameter of the ring distribution of every protein, going from the smallest one, SAS-6[N], to the most important ones, SAS-7[N], MZT-1, and TBG-1 (Fig 3C). This evaluation established that almost all elements that have been proven beforehand by biochemical and cell organic assays to bodily work together are certainly situated in shut neighborhood to 1 one other. That is the case for SAS-6 and SAS-5 [43], SAS-4 and HYLS-1 [17], SAS-7 and SPD-2 [8,44], SPD-2 and SPD-5 [45], in addition to PCMD-1 and SAS-4 or SPD-5 [46].

Total, U-Ex-STED enabled us to localize in a complete method centriolar and PCM core element with unprecedented spatial precision.

9-fold symmetrical and chirality establishing elements of the C. elegans centriole

We subsequent addressed whether or not the ring-like distribution of every centriolar and PCM core element reveals 9-fold radial symmetry. To this finish, we carried out an evaluation of the U-Ex-STED information set for every element that’s illustrated within the case of α-tubulin in Fig 4A and 4B. First, a circle was drawn alongside the ring-like sign and an depth profile measured alongside this round line (Fig 4A and 4B). Within the majority of circumstances, this yielded 9 clearly distinguishable peaks. In ultimate prime views, with no or little or no tilt of the organelle with respect to the imaging axis, the common distance between sign peaks is in line with the 40° angle anticipated from a 9-fold radially symmetric construction (Fig 4C). Importantly, in addition to α-tubulin, we discovered a 9-fold symmetric association for SPD-5, PCDM-1[C], SPD-2[C], HYLS-1[N], SAS-4, SAS-6[C], and SAS-1[N] (Fig 4D–4K, left two panels, uncooked).

thumbnail

Fig 4. Chiral options of the centriole.

(A) U-Ex-STED of a centriole stained for α-tubulin. Numbers correspond to the sign peaks within the depth profile alongside the dashed line reported in (B). (B) Sign depth profile alongside the dashed line in (A) (4 pixels broad). (C) Angles between peaks of α-tubulin sign depth profiles in 14 prime views of centrioles imaged with U-Ex-STED. Angles have been decided by dividing the gap between every neighboring peak by the size of the complete profile, multiplied by 360. Alternating gray and black information factors point out values from every of the 14 centrioles examined. 11/14 centrioles confirmed 9 clearly discernable sign peaks, 2/14 solely 8, and 1/14 apparently 10 such peaks. (D-Okay) U-Ex-STED and plot sign depth profiles of uncooked pictures (left two panels) and corresponding 9-fold symmetrized variations (proper) of the indicated pairs of elements (inexperienced and magenta). In circumstances prime views have been barely tilted, pictures have been circularized earlier than 9-fold symmetrization utilizing the Fiji plugin “Rework-Interactive Affine”. The numbers on the very proper signify the common offset of the two nearest sign depth peaks in 9-fold symmetrized pictures for the two elements examined. (L) (Left) U-Ex-STED of centriole from a worm expressing SPD-2::GFP stained for GFP and α-tubulin in the identical shade (inexperienced), in addition to for SAS-4 (magenta). (Center) Corresponding 9-fold symmetrized model. (Proper) Schematic illustration of the IF evaluation, manually separating the SPD-2::GFP and α-tubulin indicators primarily based on their ring diameter. Purple bins are magnified on the correct of every picture. Be aware that we’ve not analyzed HYLS-1 on this method, because the mCherry::HYLS-1 sign depth was too weak to this finish. Knowledge underlying the graphs proven within the determine may be present in S1 Knowledge. IF, immunofluorescence; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.


https://doi.org/10.1371/journal.pbio.3001784.g004

We subsequent investigated whether or not centriolar proteins thus localized exhibit an offset distribution with respect to microtubules. To that finish, we examined if the 9-fold radial symmetric distributions are on the identical angular axis because the microtubules utilizing the next evaluation pipeline. First, we averaged the indicators of the microtubules and of the element to be examined by making use of 9-fold symmetrization (S2 Fig; [47,48]). Second, we acquired a sign depth plot alongside the ring within the ensuing symmetrized pictures for each channels. As anticipated, given the 9-fold symmetrization, in such an evaluation particular person sign peaks for every channel are roughly 40° aside (360°/9 sign peaks) (Fig 4D–4K). Third, sign depth plots from the two channels are overlaid, and the angular distance between every peak within the α-tubulin channel and the neighboring peak within the second channel decided. On this method, the common angular offset in every centriole of the element in query versus microtubules is computed.

Strikingly, the above evaluation pipeline revealed that elements exhibiting a 9-fold symmetric association fall into 2 well-separated teams. In a primary two-membered group containing SPD-5 and PCMD-1[C], the offset with respect to microtubules is marginal (<3°) (Fig 4D and 4E), much like that of the two antibodies raised towards α-tubulin (2.4° ± 1.6, N = 10). Subsequently, SPD-5 and PCMD-1[C] aren’t offset with respect to the microtubules. In stark distinction, a second group of elements exhibited a transparent offset (>9°) with respect to the microtubules, thus resulting in a chiral ensemble (Fig 4F–4K). This second group contains SPD-2[C] and HYLS-1[N], that are each situated exterior the microtubules (Fig 4F and 4G). As well as, SAS-4 and SAS-1[N], which each have a hoop diameter much like that of α-tubulin, exhibited an offset with respect to microtubules (Figs 3C and 4H). Extra internally, SAS-6[C] additionally reveals a robust offset with respect to microtubules (Fig 4I). Importantly, we discovered moreover that SAS-4 and SAS-6[C] are effectively aligned with each other (Fig 4J), as are SAS-4 and SAS-1[N] (Fig 4K). Thus, the offset of SAS-4 reveals the identical handedness with respect to microtubules as that of SAS-6 and SAS-1.

How does the offset handedness of elements situated exterior the microtubules relate to that of these situated extra centrally? To handle this query, we got down to concurrently look at offset within the angular axis with respect to microtubules of the outer elements SPD-2[C] and the interior offset element SAS-4. As a result of our microscopy setup doesn’t lend itself to performing high-quality 3-color STED, we marked SPD-2[C] and α-tubulin in the identical shade on this experiment, since they exhibit clearly distinct ring diameters (see Fig 3A and 3C, #8 and #14). Importantly, this evaluation uncovered that SPD-2[C] and SAS-4 are invariably situated on the identical facet of the microtubules (Fig 4L). Be aware that that is no matter whether or not SPD-2[C] and SAS-4 are to the left or to the correct of the α-tubulin sign, which is anticipated to rely upon whether or not a centriole is seen from the proximal or the distal finish. We conclude that the offset of the outer element SPD-2[C] and the interior element SAS-4 has the identical handedness with respect to microtubules.

Ultrastructural map of the C. elegans gonad centriole

Having achieved exact protein localizations within the gonad centriole with U-Ex-STED, we got down to decide whether or not we are able to assign particular centriolar elements to particular compartments of the organelle. Provided that the ultrastructure of the worm centriole has been greatest studied within the early embryo [68], and contemplating that the centriole might exhibit tissue-specific options, we got down to straight characterize the ultrastructure of the gonad centriole. Utilizing Correlative Gentle Electron Microscopy (CLEM) of chemically mounted samples to ease recognizing of G2 early meiotic prophase centrioles adopted by 50 nm serial sectioning, we acquired the most important EM information set of worm centrioles to this point (N = 44). As proven in Fig 5A and 5B, in addition to in S3A Fig, we discovered that peripheral paddlewheels, microtubules, central tube, and interior tube are all clearly discernable, as they’re within the early embryo [68]. Nonetheless, facet views established that the centriole is shorter within the gonad than within the early embryo (96.6 ± 7.2 nm as in comparison with roughly 175 nm [7,8]). Furthermore, prime view revealed that the paddlewheels are barely smaller as effectively [8]. Aside from these 2 variations, we conclude that the general ultrastructure of the centriole is conserved between the embryo and the gonad.

thumbnail

Fig 5. EM evaluation of the centriole within the gonad.

(A) (Left) EM of prime and facet views, as indicated, of early meiotic prophase centrioles. (Proper) Overlay with distinct ultrastructural compartments as described in (C). (B) Diameters of ultrastructural compartments of the gonad centriole. High view (crosses) and facet views (circles) have been analyzed; mild and darkish shade of colours signify information factors from 2 unbiased samples. The center strains of the boxplots correspond to the median, the field contains 50% of values (IQR), and the whiskers present the vary of values inside 1.5*IQR. (C) Schematic illustration of prime and facet views of centrioles with ultrastructural compartments depicted in colours, in addition to common measurements ± SD (see additionally C). N = 38 for centriole top. Knowledge underlying the graphs proven within the determine may be present in S1 Knowledge. EM, electron microscopy; SD, customary deviation.


https://doi.org/10.1371/journal.pbio.3001784.g005

Utilizing each prime and facet views, we decided the diameter of the complete organelle, encompassing probably the most peripheral paddlewheel options, to be 140.0 ± 9.5 nm (Fig 5B and 5C). Furthermore, the diameters of central tube and interior tube are 51.5 ± 2.5 nm and 28.6 ± 2.3 nm, respectively, whereas that of the radial array of the microtubules is 87.9 ± 5.7 nm (Fig 5C). These measurements are in step with these from high-pressure frozen early embryonic centrioles [7,8], indicating that there isn’t any or marginal shrinkage on account of chemical fixation.

The paddlewheels of the centriole within the embryo have been reported to exhibit a clockwise twist with respect to the microtubules when seen from the distal finish, utilizing the presence of the procentriole to outline the proximal finish of the centriole [8]. Within the gonad, nevertheless, the place the centriole is shorter, the procentriole usually seems to cowl the centriole top in its entirety (S3B Fig; N = 15). In these circumstances the place the centriole is larger than the cross-sectional diameter of the procentriole (N = 17), the latter might typically emanate from the neighborhood of one of many 2 ends (S3C Fig). The observations made by EM have been supported and complemented by analyses carried out with U-Ex STED. Thus, we discovered that when the centriole is brief, the procentriole emanates from its whole top (S3D Fig, left). When the centriole is larger, the procentriole may be both centered with respect to the centriole (S3D Fig, center) or positioned nearer to 1 finish (S3D Fig, proper). Subsequently, we speculate that, in any case within the gonad, the procentriole can emanate from any web site alongside the pourtour of the centriole. Regardless, chirality of the centriole can’t be assessed reliably with respect to procentriole orientation within the gonad. Nonetheless, the truth that the procentriole can even emanate from the center of the centriole raises the likelihood that centriole chirality won’t be mounted with respect to procentriole orientation.

Establishing the molecular structure of the C. elegans centriole: Past microtubules

To higher perceive the mobile context wherein the centriole resides, we carried out tomographic evaluation of the EM sections (ET), which revealed a ribosome free space roughly 262 ± 26 nm in diameter extending past the paddlewheels (Fig 6A; N = 3). This diameter is roughly 60 nm bigger than that of the most important ring-like distribution noticed on this work (see Fig 3C), elevating the likelihood that different proteins could also be current on this space.

thumbnail

Fig 6. Overlay of EM and U-Ex pictures.

(A) (Left) Max depth Z-projection of ET of an early meiotic prophase centriole and surrounding area. (Center) Magnification of the black field within the picture on the left. (Proper) Manually annotated ribosomes are proven in magenta and paddlewheel constructions with dark-yellow outlines. Be aware that the ribosome-free space extends past the paddlewheels. Purple inset reveals a magnified ribosome from the identical ET picture. (B, C, F, G) Overlay of U-Ex-STED and EM pictures (inverted gray ranges) of centrioles from early meiotic prophase. Circularized pictures (left two panels), corresponding 9-fold symmetrized variations (subsequent two panels), and magnification of the insets highlighted by the white field (very proper). (B) Be aware that SAS-7 extends past the paddlewheel. (C) Overlay of paddlewheel elements. (F) Overlay of elements round microtubules. (G) Overlay of SAS-6 (N- and C-ter) and microtubules. (D, E) (Left) Magnification of a 9-fold symmetrized centriole imaged by EM (D) and highest populated class from class-averaging of particles containing microtubules and paddlewheels from particular person ET tilt collection of 4 centrioles (E) (see S4 Fig). Photos are colorized with the LUT “Hearth” (low intensities in blue, excessive intensities in magenta and purple). Gentle inexperienced arrowheads level to the small density subsequent to the paddlewheel (IPD), stuffed white arrowheads to that spanning from the central tube to 1 facet of the microtubule (SCD). (Proper) Depth profiles have been obtained alongside the indicated dashed strains (10 pixels broad). Microtubules show constantly extra density on the facet situated underneath the paddlewheel (marked with an A) in comparison with the opposite facet (marked with a B). EM, electron microscopy; ET, electron tomography; IPD, Inter Paddlewheel Density; SCD, SAS-6/4/1 Containing Density; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.


https://doi.org/10.1371/journal.pbio.3001784.g006

We got down to decide the id of the centriolar and PCM core proteins that correspond to given ultrastructural compartment of the organelle. To this finish, we devised a technique that depends on overlaying U-Ex-STED and EM pictures, utilizing microtubules as a joint registration customary. Briefly, we circularized, rotated, and size-adjusted collectively the two U-Ex-STED channel indicators, aligning the α-tubulin sign with the microtubules within the EM pictures (S4 Fig). We utilized this technique initially on the symmetrized pictures after which likewise adjusted the uncooked information (S4 Fig). We report the outcomes of this evaluation hereafter, beginning with the surface of the organelle.

Overlaying the U-Ex-STED and EM information revealed that SAS-7[N] localizes simply exterior the paddlewheel, partially filling the area devoid of ribosomes surrounding the centriole (Fig 6B). 4 elements have been discovered to localize to the paddlewheel: HYLS-1[N], SPD-2, SPD-5, and PCMD-1. SPD-5 and PCMD-1[C] are on the identical angular axis as microtubules within the U-Ex-STED information set (see Fig 4D and 4E), and we certainly discover PCMD-1[C] simply outwards of microtubules within the overlay, constituting the bottom of the paddlewheel (Fig 6C). HYLS-1[N] additionally localizes to the bottom of the paddlewheel, however in distinction to SPD-5 and PCMD-1[C], it does so with an offset with respect to the microtubules (Fig 6C). SPD-2 is the outermost element of the paddlewheel with the two ends exhibiting distinct distributions: SPD-2[C] seems as foci positioned simply exterior of microtubules, with an angular offset with respect to them (Fig 6C; see additionally Fig 4F), whereas SPD-2[N] localizes barely additional to the surface as an epitrochoid with 9 lobes extending left and proper over the paddlewheel (Fig 6C). Curiously, we detected a beforehand unnoticed small electron-dense area within the EM and ET information units (see under) situated between neighboring paddlewheels (Fig 6D and 6E, inexperienced arrowheads), which may be partially matched with the place of SPD-2[N] in these overlays. We title this density Inter Paddlewheel Density (IPD). Total, this evaluation reveals in beautiful element the molecular structure of elements situated exterior the centriolar microtubules.

Molecular structure on the stage of the microtubules

We subsequent report the evaluation of elements situated extra centrally. Upon cautious evaluation of the symmetrized EM information set, we seen one other novel density, which begins from the central tube (Fig 6D, dashed arrowhead), extends in the direction of and alongside every microtubule, rendering 1 facet of the microtubule extra pronounced than the opposite (Fig 6D, white arrowhead). This density shows the identical angular offset with respect to the microtubules because the paddlewheel. Since microtubules aren’t at all times completely perpendicular to the airplane of sectioning, we carried out ET to acquire bona fide prime views of microtubules and thus higher analyze this novel density. From particular person tilt collection of 4 centrioles, we picked 628 particles containing microtubules and paddlewheels; class-averaging resulted in 3 well-defined lessons containing 92% of enter particles (S5A Fig). In all 3 lessons, the novel density is current on the facet of the microtubule above which the paddlewheel is situated (Figs 6E and S5B). Provided that SAS-6, SAS-4, and SAS-1 all show the identical angular offset route with respect to microtubules because the paddlewheel element SPD-2[C] (see Fig 4F and 4H–4L), we suggest that these 3 proteins collectively might compose this novel offset density. Subsequently, we title this novel density “SAS-6/4/1 Containing Density” (SCD). Overlays of the corresponding U-Ex-STED and EM pictures certainly revealed good alignment of SAS-4, SAS-6[C], and SAS-1[N] with the SCD, under 1 facet of the microtubule (Figs 6F, 6G, and S5C). Furthermore, SAS-4[C] overlaps virtually completely with the SCD on the stage of the central tube, whereas SAS-1[N] has an indistinguishable diameter from SAS-6[C] (see Fig 3C). Taken collectively, our information counsel that SAS-4, SAS-6, and SAS-1 kind the newly described chiral SCD, with SAS-4 probably bridging it to HYLS-1.

The N-terminus of SAS-6 is current on the interior tube and doesn’t kind a spiral

We capitalized on the unprecedented excessive decision afforded by U-Ex-STED to handle whether or not C. elegans SAS-6 varieties a hoop or as a substitute a steep spiral in vivo, as has been hypothesized primarily based on structural and biophysical information [16]. The spiral mannequin predicts that SAS-6[N] ought to be obvious in prime views as a small ring with a diameter of roughly 4.5 nm [16]. Given the roughly 14 nm efficient lateral decision achieved utilizing U-Ex-STED, this would seem as a single focus. Opposite to this prediction, we discovered that the diameter of the ring shaped by SAS-6[N] is roughly 31 ± 3 nm, overlapping with the interior tube in EM pictures (Fig 6G). We famous additionally that SAS-6[C] localizes roughly 41 ± 4 nm away from SAS-6[N], in step with the truth that the coiled-coil area of SAS-6 is roughly 35 nm lengthy and adopted by an intrinsically disordered area of roughly 90 amino acids [49]. Taken collectively, our observations point out that, slightly than a steep spiral, in vivo, C. elegans SAS-6 varieties a ring-containing cartwheel.

Dialogue

We deciphered the molecular structure of the minute C. elegans centriole in unprecedented element by combining U-Ex-STED with EM, thereby localizing 12 centriolar and PCM core proteins to distinct ultrastructural compartments (Fig 7).

thumbnail

Fig 7. Schematic illustration of the localization of elements inside the centriole.

(High left) 9-fold symmetrized EM picture of a centriole. (High center) Overlay of the picture on the left with corresponding schematic illustration on the correct. (High proper) Coarse grained schematic illustration of the localization inside the centriole of elements analyzed on this research. (Backside left) Magnification of the blue boxed area from the picture above. Inexperienced field highlights the IPD, white field the SCD. Dashed circle highlights a small density to which no identified centriolar protein has been assigned (see Dialogue). (Backside center and proper) Magnifications of the blue boxed areas from the photographs above, with a sign of the localization of every protein, together with the place the N- and C-terminus maps when identified. No point out of termini is indicated when merely antibodies have been utilized. EM, electron microscopy; IPD, Inter Paddlewheel Density; SCD, SAS-6/4/1 Containing Density.


https://doi.org/10.1371/journal.pbio.3001784.g007

The exact localization of proteins achieved herein is by and huge suitable with, and extends, earlier findings. Thus, elements that have been proven beforehand by biochemical and cell organic assays to bodily work together are certainly situated in shut neighborhood of each other in our map, together with SAS-6 and SAS-5 [43], SAS-4 and HYLS-1 [17], SAS-7 and SPD-2 [8,44], SPD-2 and SPD-5 [45], in addition to PCMD-1 with SAS-4 and SPD-5 [46]. Different distributions weren’t essentially anticipated from prior work. As an illustration, we discovered that SAS-7 localizes partly exterior the paddlewheel construction, inside the zone of ribosome exclusion. SPD-2 and PCMD-1 aren’t wanted for SAS-7 localization, whereas SAS-7 is required for regular centriolar ranges of SPD-2 and PCMD-1, in addition to for integrity of the paddlewheels themselves [8,46]. The localization of SAS-7 exterior of SPD-2 and PCMD-1 raises the likelihood that SAS-7 capabilities by a shielding mechanism slightly than by recruiting SPD-2 and PCMD-1. One other functionally suggestive distribution uncovered right here is that of SAS-1: SAS-1[N] localizes simply contained in the microtubule ring, in step with the truth that this a part of the protein associates with and stabilizes microtubules when ectopically expressed in human cells [18]. Subsequently, it’s tempting to invest that C. elegans SAS-1 maintains centriole integrity by domestically exerting a microtubule stabilizing operate. The one reported interplay not recapitulated right here is that of microtubules with SAS-5, which is mediated by its N-terminus and lead to colocalization with the microtubule community upon transfection in COS-7 cells [50]. We used antibodies raised towards this area of SAS-5 and located them to localize roughly 50 nm extra centrally than the centriolar microtubules, suggesting that the SAS-5 N-terminus interacts preferentially with one other element within the C. elegans centriole.

Overlaying EM pictures with U-Ex-STED pictures of all identified centriolar proteins to this point permits us to contemplate whether or not there could also be centriolar proteins that haven’t but been recognized within the worm. Such proteins may correspond to EM densities that can not be readily accounted by the distribution of the proteins assessed right here by U-Ex-STED. One such density, distinct from the ICP and the SCD, is obvious on the interior facet of microtubules, reverse the SCD (see Fig 7, dashed circle). That is harking back to the area to which Cep135 and Cep97 localize within the fly centriole [34]. It has been steered {that a} divergent Cep135 protein localizes to centrioles of C. elegans throughout sure developmental levels [51], and it will likely be attention-grabbing to make use of U-Ex-STED to handle whether or not it maps to this density. Alternatively, it’s doable {that a} phase of the proteins examined right here and localizing to such a density wouldn’t be obvious from tagging the N- or C-terminal a part of the protein, or with among the antibodies that have been utilized.

Our method enabled us to probe the upper order oligomerization mechanisms of SAS-6 in vivo. Earlier structural and biophysical experiments led to the suggestion that such oligomers kind a steep spiral as a substitute of a hoop as in different programs [16]. The steep spiral mannequin predicts that SAS-6[N] seems in prime views as a small ring with a diameter of roughly 4.5 nm [16].

We discovered as a substitute that SAS-6[N] varieties a hoop roughly 31 ± 3 nm in diameter, which neatly overlays with the interior tube ultrastructural compartment in EM pictures. We conclude that SAS-6 doesn’t kind a steep spiral within the worm and suggest as a substitute that the protein assembles right into a ring-containing cartwheel as in different organisms. Alternatively, C. elegans SAS-6 might assemble right into a shallow spiral. Furthermore, we discovered SAS-6[C] to be positioned roughly 41 ± 4 nm away from SAS-6[N], suitable with a cartwheel construction wherein the SAS-6 coil-coil domains kind spokes extending in the direction of the peripheral microtubules, as in different programs [14,15]. Will probably be attention-grabbing to uncover how the intrinsic properties of C. elegans SAS-6 that allow it to kind a steep spiral in vitro are modulated within the organismal context to undertake a ring-like configuration. This is likely to be aided by interacting proteins, akin to SAS-5 [44,50,52], or by a connection of SAS-6[C] to microtubules. Alternatively, properties of the centriole floor from which the procentriole assembles may impose a special conformation, for the reason that presence of a floor may help constrain the inherent helical properties of Chlamydomonas reinhardtii SAS-6 polymers into a hoop [53].

Our evaluation uncovered offset protein distributions with respect to microtubules, thereby leading to a chiral centriolar ensemble. Such an offset pertains notably to SAS-6[C] and SAS-4, which coincide with the newly recognized SCD, an EM density discovered centrally and laterally to the microtubule wall. Curiously, the SCD shows an angular offset with respect to the microtubule in the identical route because the paddlewheel and its constituent SPD-2[C]. Angular offsets with respect to centriolar microtubules happen in different programs. For instance, EM research of centrioles in Trichonympha and Chlamydomonas uncovered that the pinhead element connecting cartwheel and microtubules reveals an angular offset with respect to the A microtubule that’s on the facet of the B and C microtubules [54]. Furthermore, super-resolution microscopy in Drosophila revealed that the centriolar proteins Cep135 and Ana1 exhibit an angular offset with respect to the A microtubule that’s situated on the facet reverse the B microtubule [34].

Chirality of the centriole is a signature function of the organelle noticed throughout the eukaryotic area of life. Nonetheless, the potential evolutionary stress resulting in conservation of such chirality isn’t clear, though an interesting risk is that this might be optimum for ciliary and flagellar motility. Regardless, it has been steered that centriolar chirality could also be imparted by inherent chiral options of SAS-6 proteins, with chirality within the interior a part of the organelle dictating that of extra peripheral components, together with microtubules [53]. Alternatively, chirality might stem from the truth that microtubules of the procentriole develop with a set orientation from the floor of the centriole, with the plus finish main. Subsequently, the surfaces for molecular interplay accessible on the left and the correct facet of a microtubule are inherently completely different. Because of this, a protein that interacts with a particular floor on the microtubule wall and that has a set orientation alongside the polymer, akin to SAS-4 (see Fig 7), would essentially render the centriole chiral. Regardless, it will likely be essential to uncover how chirality of the centriole is established and what its position is likely to be in centriolar biogenesis and performance.

Will probably be of curiosity to use the strategies developed herein to probe potential variations within the molecular structure of centrioles in distinct developmental contexts in C. elegans. For instance, we uncovered right here that centriole size within the gonad and the early embryo differ considerably; such a top distinction could also be accompanied by alterations in molecular structure. Furthermore, these strategies may be deployed to interrogate with utmost precision the molecular structure of centrioles in mutant worms on this genetically tractable organism to additional unravel mechanisms of organelle biogenesis and performance. Past C. elegans, such an analytical framework is anticipated to likewise reveal the distribution of centriolar proteins in different programs, and thereby determine conserved and variable options of organelle structure.

Supplies and strategies

C. elegans tradition situations

Worms have been grown on E. coli (OP50) seeded NG agar plates at 20°C and age matched as L1 larvae by bleaching gravid adults in accordance with [63]. Worms have been harvested for ethanol fixation or gonad spreading 24 to 36 hours post-L4 stage by washing them off the plate with PBS-T (PBS equipped with 0.1% Tween-20).

CRISPR/Cas9 genome modifying

3xFLAG tagging of SAS-1 and SAS-6 was carried out by CRISPR/Cas9 as described in [64]. crRNAs have been designed utilizing the GUIDE DESIGN instrument (http://crispr.mit.edu). Briefly, younger grownup worms have been injected with CRISPR/Cas9 ribonuclear complexes (selfmade, 2.5 μg/μl), and dpy-10 was used because the coinjection marker. ssDNA restore templates have been injected at concentrations of 414 nM for dpy-10 and 500 nM for the 3xFlag tags, respectively. F1 progenies with curler or dumpy phenotypes have been chosen and the edits assessed utilizing PCR within the F2 technology, adopted by verification by Sanger sequencing.

The crRNA sequences have been as follows:

SAS-1 N-terminus: ACAATTACTGGTGCCCTTCT(CGG) (30 μM)

SAS-1 C-terminus: CGGATTTGGAGAATATGATG(AGG) (30 μM)

SAS-6 N-terminus: AATTTTGCTAGTCATTTTTG(TGG) (30 μM)

Gonad spreading

Spreading of C. elegans gonads was carried out in the same method as in [39]. Gonads of roughly 1,000 grownup worms have been dissected in 30 μL dissection resolution (0.2 × PBS, 1:1,000 Tween 20) on an ethanol-washed 22 × 40 mm coverslip. A quantity of 5 to 10 μL of dissected gonads have been then pipetted onto a brand new ethanol-washed 22 × 40 mm coverslip, and 50 μL of spreading resolution (for 1 coverslip, 50 μL: 32 μL of Fixative (4% w/v Paraformaldehyde and three.2% w/v Sucrose in water), 16 μL of Lipsol resolution (1% v/v/ Lipsol in water), and a pair of μL of Sarcosyl resolution (1% w/v of Sarcosyl in water)) was added, and gonads have been instantly distributed over the coverslip utilizing a pipette tip. Coverslips have been left to dry at room temperature adopted by incubation at 37°C for 1 hour. Coverslips have been both processed for staining and growth or saved at −80°C. For every element analyzed, centrioles stem from a single experiment with roughly 1,000 animals and due to this fact a number of hundred hundreds nuclei, so that every centriole imaged very possible stems from a special animal.

Dedication of efficient decision

The decision of STED pictures was decided with 589 nm excitation and depletion with the 775 nm pulsed depletion laser in 10 uncooked pictures of α-tubulin utilizing the ImageJ plugin “ImageDecorrelationAnalysis” [65], which resulted in a decision estimate of 73.4 (± 7.96) nm. This decision was divided by the common growth issue of 5.2, decided by the perimeter of α-tubulin indicators within the U-Ex-STED pictures divided by the perimeter of microtubules in EM pictures. SDs of all 3 measurements have been summed up as a share of every particular person measurement (estimation of decision within the 10 pictures, measurements for perimeters of α-tubulin, and measurements of perimeters of microtubules).

CLEM evaluation

Gonads of genotype sas-7(or1940[gfp::sas-7])III; glo-1(zu931)X; itIs37[pie-1p::mCherry::H2B, unc-119(+)] or ltSi202[pVV103/ pOD1021; Pspd-2::GFP::SPD-5 RNAiresistant;cb-unc-119(+)]II; sas-7(is1[tagRFP::sas-7+loxP])III; glo-1(zu931)X have been dissected in sperm buffer (50 mM Hepes (pH 7.0), 50 mM NACL, 25 mM KCL, 5 mM CaCl2, 1 mM MgSO4, 50 mM Glucose, 1 mg/ml BSA) and transferred on poly-lysine-coated MatTek glass backside dishes. 3D imaging of gonads was carried out utilizing a Nikon Ti2-E epifluorescent microscope geared up with an Andor digital camera Zyla-4.2P-CL10 earlier than and after an roughly 2-hour and 30-minute fixation at room temperature in 2.2% glutaraldehyde, 0.9% Paraformaldehyde in Cacodylate buffer 0.05M (pH 7.4), 0.09 M sucrose, and 0.9 mM MgCl2. Briefly, specimens have been postfixed in 1% osmium tetroxide, 0.8% potassium ferrocyanide in cacodylate buffer (0.1 M, pH 7.2), handled with 0.2% Tannic Acid in 0.05 M cocadylate buffer (pH 7.0), stained with 1% uranyl acetate in Sodium Acetate (pH 5.2), dehydrated in an alcohol collection, and embedded in Laborious EPON. At 23,000× magnification, 50-nm sections have been imaged utilizing a TecnaiSpirit (FEI Firm) operated at 80 kV and geared up with an Eagle CCD digital camera (FEI Firm). Utilizing relative positioning of centrioles and nuclei in fluorescence pictures facilitated the search of centrioles and restricted the variety of sections to be imaged. Gaussian blur filtering 1.5 was utilized on displayed EM pictures.

Ultrastructural compartments of the centriole have been measured manually utilizing Fiji [66]. Every information level is the common of 4 measurements extracted from strains drawn alongside the peak of the function. In some circumstances, ultrastructural compartments couldn’t be measured as a result of they weren’t visualized precisely, or the view of the centriole was too tilted. Graphs have been generated utilizing PlotsofData [67] and SuperPlotsofdata [68]. Procentriole positioning relative to the centriole in S3 Fig was qualitatively assessed on facet views, excluding views that have been too tilted.

Electron tomography

Tilt-series from cryo-fixed sections have been acquired on a Tecnai F20 operated at 200 kV (Thermo Fischer Scientific utilizing Thermo Scientific Tomography software program in steady tilt scheme from −60° to +60° in 2° steps at −2.5 μm defocus. Knowledge have been recorded with a Falcon III DD digital camera (Thermo Fisher Scientific) in linear mode at 2,000× magnification, similar to a pixel dimension of three.49. Particles have been picked from particular person tilt pictures and 2D Class averages have been calculated utilizing Relion [69], Xmipp [70], and Eman2 inside the Scipion3 [71] framework. Tilt collection alignment and tomogram reconstruction was executed utilizing EMAN 2.9 [72]. Tomogram subvolumes for the detection of ribosome-free space have been extracted utilizing Imod 4.9 [73] and most depth venture in Fiji [66].

Worm strains used on this research:

  • N2 (Bristol)
  • TMD101: pcmd-1(t3421); mikSi6[pmai-2:GFP::C17D12.7] II [21]
  • TMD117: pcmd-1(t3421); mikSi9[pmai-2:C17D12.7::GFP] II [21]
  • DAM276: ltSi40 [pOD1227; Psas-6::sas-6reencoded::GFP; cb unc-119(+)] II; sas-6(ok2554) IV [57]
  • GZ1934: sas-1(is7[3xflag::sas-1]) III (this research)
  • GZ1966: sas-1(is6[sas-1::3xflag]) III (this research)
  • OC994: sas-4(bs195[sas-4::gfp] III (a present from Kevin O’Connell)
  • DAM307: vieSi16[pAD390; Phyls1:mcherry::hyls-1; cb unc-119(+)] IV [60]
  • GZ1528: spd-2(is2[tagRFP::spd-2 +loxP]) I; sas-7(or1940(gfp::sas-7)) III; glo-1(zu931) X (this research)
  • DAM640: spd-2(vie4[spd-2::gfp +loxP]) I [61]
  • JLF375: mzt-1(wow51[GFP:MZT-1]) I; zif-1(gk117) III [27]
  • GZ1929: sas-6(is10[3xflag::sas-6]) IV (this research)

Supporting data

S1 Fig. Procentriole composition and maturation.

(A) Widefield picture of two pairs of centriole/procentriole in an S-phase cell situated within the mitotic zone of the gonad after U-Ex, stained for SAS-6 and α-tubulin. Be aware that procentrioles harbor SAS-6 however no α-tubulin at this stage. (B) U-Ex-STED pictures of centrioles from meiotic prophase (left) and the mitotic zone (center and proper). Photos illustrate that the quantity of ZYG-1 on the centriole (however not on the procentriole) varies: Throughout meiotic prophase, ZYG-1 ranges on the centriole are very low. For quantification, a line was drawn from the middle of the centriole to the surface of the procentriole and the depth profile alongside this line measured, as represented by the dashed arrows. (Left) In 95% of meiotic prophase centrioles (19/20), a single ZYG-1 sign depth peak was detected exterior of the centriole peak, straight underneath the procentriole. (Center and proper) Within the mitotic zone, against this, whereas 12/25 centrioles exhibited the same distribution to that noticed within the overwhelming majority of meiotic prophase centrioles, 13/25 displayed the ZYG-1 sign extra prominently than throughout meiotic prophase, with 2 clear ZYG-1 peaks, one on the base of the procentriole and one in the course of the centriolar SAS-6::GFP sign. (C) 3D-SIM sum depth Z-projected picture of a nucleus within the mitotic zone of an expanded gonad. Phosphorylated Histone 3 marks nuclei in mitosis. Insets on the correct present that every one 4 centrioles include α-tubulin, in contrast to in S or G2 section. SIM, Structured Illumination Microscopy; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.

https://doi.org/10.1371/journal.pbio.3001784.s001

(PDF)

S4 Fig. Schematic of U-Ex and EM pictures picture overlay.

In EM and U-Ex-STED prime views, centrioles with slight tilted orientations have been circularized with the Fiji plugin “Rework-Interactive Affine”. The gray ranges of the EM picture have been inverted, and the circularized EM and U-Ex-STED pictures then 9-fold symmetrized as illustrated in S2 Fig. The sides of the microtubule wall in symmetrized EM pictures and of the α-tubulin sign in symmetrized U-Ex-STED pictures have been measured, and the symmetrized U-Ex-STED picture adjusted in dimensions in order that the α-tubulin sign had the identical perimeter because the microtubule wall in symmetrized EM. To overlay symmetrized pictures, U-Ex-STED pictures have been rotated in order that particular person α-tubulin indicators completely overlapped with particular person microtubule indicators in symmetrized EM pictures. Thereafter, pictures have been overlayed in particular person shade channels. The rotational angles and dimension changes utilized for symmetrized pictures have been then utilized additionally to the uncooked nonsymmetrized pictures (indicated by the dashed arrows), which have been then handled likewise. EM, electron microscopy; U-Ex-STED, Ultrastructure Growth coupled with STimulated Emission Depletion.

https://doi.org/10.1371/journal.pbio.3001784.s004

(PDF)

References

  1. 1.
    Azimzadeh J. Exploring the evolutionary historical past of centrosomes. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130453. pmid:25047607
  2. 2.
    Bornens M. The centrosome in cells and organisms. Science. 2012;335:422–426. pmid:22282802
  3. 3.
    Winey M, O’Toole E. Centriole construction. Philos Trans R Soc B Biol Sci. 2014;369:20130457. pmid:25047611
  4. 4.
    Uzbekov R, Prigent C. Clockwise or anticlockwise? Turning the centriole triplets in the correct route! FEBS Lett. 2007;581:1251–1254. pmid:17368628
  5. 5.
    Ito D, Bettencourt-Dias M. Centrosome Remodelling in Evolution. Cell. 2018;7:E71. pmid:29986477
  6. 6.
    O’Toole ET, McDonald KL, Mäntler J, McIntosh JR, Hyman AA, Müller-Reichert T. Morphologically distinct microtubule ends within the mitotic centrosome of Caenorhabditis elegans. J Cell Biol. 2003;163:451–456. pmid:14610052
  7. 7.
    Pelletier L, O’Toole E, Schwager A, Hyman AA, Müller-Reichert T. Centriole meeting in Caenorhabditis elegans. Nature. 2006;444:619–623. pmid:17136092
  8. 8.
    Sugioka Okay, Hamill DR, Lowry JB, McNeely ME, Enrick M, Richter AC, et al. Centriolar SAS-7 acts upstream of SPD-2 to control centriole meeting and pericentriolar materials formation. Elife. 2017;6:e20353. pmid:28092264
  9. 9.
    Wolf N, Hirsh D, McIntosh JR. Spermatogenesis in males of the free-living nematode, Caenorhabditis elegans. J Ultrastruct Res. 1978;63:155–169. pmid:671581
  10. 10.
    Banterle N, Gönczy P. Centriole Biogenesis: From Figuring out the Characters to Understanding the Plot. Annu Rev Cell Dev Biol. 2017;33:23–49. pmid:28813178
  11. 11.
    Nigg EA, Holland AJ. As soon as and solely as soon as: mechanisms of centriole duplication and their deregulation in illness. Nat Rev Mol Cell Biol. 2018;19:297–312. pmid:29363672
  12. 12.
    Gomes Pereira S, Dias Louro MA, Bettencourt-Dias M. Biophysical and Quantitative Ideas of Centrosome Biogenesis and Construction. Annu Rev Cell Dev Biol. 2021;37:43–63. pmid:34314592
  13. 13.
    Delattre M, Canard C, Gönczy P. Sequential protein recruitment in C. elegans centriole formation. Curr Biol. 2006;16:1844–1849. pmid:16979563
  14. 14.
    Vakonakis I. The centriolar cartwheel construction: symmetric, stacked, and polarized. Curr Opin Struct Biol. 2021;66:1–7. pmid:32956907
  15. 15.
    Guichard P, Hamel V, Gönczy P. The Rise of the Cartwheel: Seeding the Centriole Organelle. BioEssays Information Rev Mol Cell Dev Biol. 2018;40:e1700241. pmid:29508910
  16. 16.
    Hilbert M, Erat MC, Hachet V, Guichard P, Clean ID, Flückiger I, et al. Caenorhabditis elegans centriolar protein SAS-6 varieties a spiral that’s in line with imparting a ninefold symmetry. Proc Natl Acad Sci U S A. 2013;110:11373–11378. pmid:23798409
  17. 17.
    Dammermann A, Pemble H, Mitchell BJ, McLeod I, Yates JR, Kintner C, et al. The hydrolethalus syndrome protein HYLS-1 hyperlinks core centriole construction to cilia formation. Genes Dev. 2009;23:2046–2059. pmid:19656802
  18. 18.
    von Tobel L, Mikeladze-Dvali T, Delattre M, Balestra FR, Blanchoud S, Finger S, et al. SAS-1 is a C2 area protein vital for centriole integrity in C. elegans. PLoS Genet. 2014;10:e1004777. pmid:25412110
  19. 19.
    Leidel S, Gönczy P. SAS-4 is important for centrosome duplication in C elegans and is recruited to daughter centrioles as soon as per cell cycle. Dev Cell. 2003;4:431–439. pmid:12636923
  20. 20.
    Pintard L, Bowerman B. Mitotic Cell Division in Caenorhabditis elegans. Genetics. 2019;211:35–73. pmid:30626640
  21. 21.
    Erpf AC, Stenzel L, Memar N, Antoniolli M, Osepashvili M, Schnabel R, et al. PCMD-1 Organizes Centrosome Matrix Meeting in C. elegans. Curr Biol. 2019;29:1324–1336.e6. pmid:30982652
  22. 22.
    Kemp CA, Kopish KR, Zipperlen P, Ahringer J, O’Connell KF. Centrosome maturation and duplication in C. elegans require the coiled-coil protein SPD-2. Dev Cell. 2004;6:511–523. pmid:15068791
  23. 23.
    Pelletier L, Ozlü N, Hannak E, Cowan C, Habermann B, Ruer M, et al. The Caenorhabditis elegans centrosomal protein SPD-2 is required for each pericentriolar materials recruitment and centriole duplication. Curr Biol. 2004;14:863–873. pmid:15186742
  24. 24.
    Hamill DR, Severson AF, Carter JC, Bowerman B. Centrosome maturation and mitotic spindle meeting in C. elegans require SPD-5, a protein with a number of coiled-coil domains. Dev Cell. 2002;3:673–684. pmid:12431374
  25. 25.
    Strome S, Powers J, Dunn M, Reese Okay, Malone CJ, White J, et al. Spindle dynamics and the position of gamma-tubulin in early Caenorhabditis elegans embryos. Mol Biol Cell. 2001;12:1751–1764. pmid:11408582
  26. 26.
    Hannak E, Oegema Okay, Kirkham M, Gönczy P, Habermann B, Hyman AA. The kinetically dominant meeting pathway for centrosomal asters in Caenorhabditis elegans is gamma-tubulin dependent. J Cell Biol. 2002;157:591–602. pmid:12011109
  27. 27.
    Sallee MD, Zonka JC, Skokan TD, Raftrey BC, Feldman JL. Tissue-specific degradation of important centrosome elements reveals distinct microtubule populations at microtubule organizing facilities. PLoS Biol. 2018;16:e2005189. pmid:30080857
  28. 28.
    Hannak E, Kirkham M, Hyman AA, Oegema Okay. Aurora-A kinase is required for centrosome maturation in Caenorhabditis elegans. J Cell Biol. 2001;155:1109–1116. pmid:11748251
  29. 29.
    Schumacher JM, Ashcroft N, Donovan PJ, Golden A. A extremely conserved centrosomal kinase, AIR-1, is required for correct cell cycle development and segregation of developmental elements in Caenorhabditis elegans embryos. Growth. 1998;125:4391–4402. pmid:9778499
  30. 30.
    Srayko M, Quintin S, Schwager A, Hyman AA. Caenorhabditis elegans TAC-1 and ZYG-9 kind a fancy that’s important for lengthy astral and spindle microtubules. Curr Biol. 2003;13:1506–1511. pmid:12956952
  31. 31.
    Bellanger JM, Gönczy P. TAC-1 and ZYG-9 kind a fancy that promotes microtubule meeting in C. elegans embryos. Curr Biol. 2003;13:1488–1498. pmid:12956950
  32. 32.
    Gartenmann L, Wainman A, Qurashi M, Kaufmann R, Schubert S, Raff JW, et al. A mixed 3D-SIM/SMLM method permits centriole proteins to be localized with a precision of ∼4–5 nm. Curr Biol. 2017;27:R1054–R1055. pmid:29017036
  33. 33.
    Yang TT, Chong WM, Wang W-J, Mazo G, Tanos B, Chen Z, et al. Tremendous-resolution structure of mammalian centriole distal appendages reveals distinct blade and matrix useful elements. Nat Commun. 2018;9:2023. pmid:29789620
  34. 34.
    Tian Y, Wei C, He J, Yan Y, Pang N, Fang X, et al. Superresolution characterization of core centriole structure. J Cell Biol. 2021;220:e202005103. pmid:33533934
  35. 35.
    Gambarotto D, Zwettler FU, Le Guennec M, Schmidt-Cernohorska M, Fortun D, Borgers S, et al. Imaging mobile ultrastructures utilizing growth microscopy (U-ExM). Nat Strategies. 2019;16:71–74. pmid:30559430
  36. 36.
    Sahabandu N, Kong D, Magidson V, Nanjundappa R, Sullenberger C, Mahjoub MR, et al. Growth microscopy for the evaluation of centrioles and cilia. J Microsc. 2019;276:145–159. pmid:31691972
  37. 37.
    Chen F, Tillberg PW, Boyden ES. Optical imaging. Growth microscopy. Science. 2015;347:543–548. pmid:25592419
  38. 38.
    Fox PM, Vought VE, Hanazawa M, Lee M-H, Maine EM, Schedl T. Cyclin E and CDK-2 regulate proliferative cell destiny and cell cycle development within the C. elegans germline. Growth. 2011;138:2223–2234. pmid:21558371
  39. 39.
    Woglar A, Yamaya Okay, Roelens B, Boettiger A, Köhler S, Villeneuve AM. Quantitative cytogenetics reveals molecular stoichiometry and longitudinal group of meiotic chromosome axes and loops. PLoS Biol. 2020;18:e3000817. pmid:32813728
  40. 40.
    Hung L-Y, Chen H-L, Chang C-W, Li B-R, Tang TK. Identification of a novel microtubule-destabilizing motif in CPAP that binds to tubulin heterodimers and inhibits microtubule meeting. Mol Biol Cell. 2004;15:2697–2706. pmid:15047868
  41. 41.
    Sharma A, Aher A, Dynes NJ, Frey D, Katrukha EA, Jaussi R, et al. Centriolar CPAP/SAS-4 Imparts Sluggish Processive Microtubule Progress. Dev Cell. 2016;37:362–376. pmid:27219064
  42. 42.
    Zheng X, Ramani A, Soni Okay, Gottardo M, Zheng S, Ming Gooi L, et al. Molecular foundation for CPAP-tubulin interplay in controlling centriolar and ciliary size. Nat Commun. 2016;7:11874. pmid:27306797
  43. 43.
    Leidel S, Delattre M, Cerutti L, Baumer Okay, Gönczy P. SAS-6 defines a protein household required for centrosome duplication in C. elegans and in human cells. Nat Cell Biol. 2005;7:115–125. pmid:15665853
  44. 44.
    Li S, Armstrong CM, Bertin N, Ge H, Milstein S, Boxem M, et al. A map of the interactome community of the metazoan C. elegans. Science. 2004;303:540–543. pmid:14704431
  45. 45.
    Boxem M, Maliga Z, Klitgord N, Li N, Lemmens I, Mana M, et al. A protein domain-based interactome community for C. elegans early embryogenesis. Cell. 2008;134:534–545. pmid:18692475
  46. 46.
    Stenzel L, Schreiner A, Zuccoli E, Üstüner S, Mehler J, Zanin E, et al. PCMD-1 bridges the centrioles and the pericentriolar materials scaffold in C. elegans. Growth. 2021;148:dev198416. pmid:34545391
  47. 47.
    Friedman MH. A reevaluation of the Markham rotation method utilizing mannequin programs. J Ultrastruct Res. 1970;32:226–236. pmid:5459067
  48. 48.
    Bezler A, Woglar A, Schneider F, Douma F, Bürgy L, Busso C, et al. Atypical and distinct microtubule radial symmetries within the centriole and the axoneme of Lecudina tuzetae. Mol Biol Cell. 2022;33:ar75. pmid:35544302
  49. 49.
    Kitagawa D, Vakonakis I, Olieric N, Hilbert M, Keller D, Olieric V, et al. Structural foundation of the 9-fold symmetry of centrioles. Cell. 2011;144:364–375. pmid:21277013
  50. 50.
    Bianchi S, Rogala KB, Dynes NJ, Hilbert M, Leidel SA, Steinmetz MO, et al. Interplay between the Caenorhabditis elegans centriolar protein SAS-5 and microtubules facilitates organelle meeting. Mol Biol Cell. 2018;29:722–735. pmid:29367435
  51. 51.
    Holzer E, Rumpf-Kienzl C, Falk S, Dammermann A. A Modified TurboID Method Identifies Tissue-Particular Centriolar Elements In C. elegans. bioRxiv. 2021:p. 2021.12.20.473533.
  52. 52.
    Qiao R, Cabral G, Lettman MM, Dammermann A, Dong G. SAS-6 coiled-coil construction and interplay with SAS-5 counsel a regulatory mechanism in C. elegans centriole meeting. EMBO J. 2012;31:4334–4347. pmid:23064147
  53. 53.
    Banterle N, Nievergelt AP, de Buhr S, Hatzopoulos GN, Brillard C, Andany S, et al. Kinetic and structural roles for the floor in guiding SAS-6 self-assembly to direct centriole structure. Nat Commun. 2021;12:6180. pmid:34702818
  54. 54.
    Guichard P, Hachet V, Majubu N, Neves A, Demurtas D, Olieric N, et al. Native structure of the centriole proximal area reveals options underlying its 9-fold radial symmetry. Curr Biol. 2013;23:1620–1628. pmid:23932403
  55. 55.
    Mikeladze-Dvali T, von Tobel L, Strnad P, Knott G, Leonhardt H, Schermelleh L, et al. Evaluation of centriole elimination throughout C. elegans oogenesis. Growth. 2012;139:1670–1679. pmid:22492357
  56. 56.
    Zhou Okay, Rolls MM, Corridor DH, Malone CJ, Hanna-Rose W. A ZYG-12–dynein interplay on the nuclear envelope defines cytoskeletal structure within the C. elegans gonad. J Cell Biol. 2009;186:229–241. pmid:19635841
  57. 57.
    Serwas D, Dammermann A. Ultrastructural evaluation of Caenorhabditis elegans cilia. Strategies Cell Biol. 2015;129:341–367. pmid:26175447
  58. 58.
    Delattre M, Leidel S, Wani Okay, Baumer Okay, Bamat J, Schnabel H, et al. Centriolar SAS-5 is required for centrosome duplication in C. elegans. Nat Cell Biol. 2004;6:656–664. pmid:15232593
  59. 59.
    Magescas J, Zonka JC, Feldman JL. A two-step mechanism for the inactivation of microtubule organizing heart operate on the centrosome. Yamashita YM, Akhmanova A, editors. Elife. 2019;8:e47867. pmid:31246171
  60. 60.
    Schouteden C, Serwas D, Palfy M, Dammermann A. The ciliary transition zone capabilities in cell adhesion however is dispensable for axoneme meeting in C. elegans. J Cell Biol. 2015;210:35–44. pmid:26124290
  61. 61.
    Laos T, Cabral G, Dammermann A. Isotropic incorporation of SPD-5 underlies centrosome meeting in C. elegans. Curr Biol. 2015;25:R648–R649. pmid:26241136
  62. 62.
    Klinkert Okay, Levernier N, Gross P, Gentili C, von Tobel L, Pierron M, et al. Aurora A depletion reveals centrosome-independent polarization mechanism in Caenorhabditis elegans. Elife. 2019;8:e44552. pmid:30801250
  63. 63.
    Stiernagle T. Upkeep of C. elegans. WormBook On-line Rev C Elegans Biol. 2006:1–11. pmid:18050451
  64. 64.
    Paix A, Folkmann A, Rasoloson D, Seydoux G. Excessive Effectivity, Homology-Directed Genome Modifying in Caenorhabditis elegans Utilizing CRISPR-Cas9 Ribonucleoprotein Complexes. Genetics. 2015;201:47–54. pmid:26187122
  65. 65.
    Descloux A, Grußmayer KS, Radenovic A. Parameter-free picture decision estimation primarily based on decorrelation evaluation. Nat Strategies. 2019;16:918–924. pmid:31451766
  66. 66.
    Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image evaluation. Nat Strategies. 2012;9:676–682. pmid:22743772
  67. 67.
    Postma M, Goedhart J. PlotsOfData-An online app for visualizing information along with their summaries. PLoS Biol. 2019;17:e3000202. pmid:30917112
  68. 68.
    Lord SJ, Velle KB, Mullins RD, Fritz-Laylin LK. SuperPlots: Speaking reproducibility and variability in cell biology. J Cell Biol. 2020;219:e202001064. pmid:32346721
  69. 69.
    Zivanov J, Nakane T, Forsberg BO, Kimanius D, Hagen WJ, Lindahl E, et al. New instruments for automated high-resolution cryo-EM construction willpower in RELION-3. Elife. 2018;7:e42166. pmid:30412051
  70. 70.
    Sorzano CO, de la Rosa Trevín JM, Otón J, Vega JJ, Cuenca J, Zaldívar-Peraza A, et al. Semiautomatic, high-throughput, high-resolution protocol for three-dimensional reconstruction of single particles in electron microscopy. Strategies Mol Biol Clifton NJ. 2013;950:171–193. pmid:23086876
  71. 71.
    de la Rosa-Trevín JM, Quintana A, Del Cano L, Zaldívar A, Foche I, Gutiérrez J, et al. Scipion: A software program framework towards integration, reproducibility and validation in 3D electron microscopy. J Struct Biol. 2016;195:93–99. pmid:27108186
  72. 72.
    Tang G, Peng L, Baldwin PR, Mann DS, Jiang W, Rees I, et al. EMAN2: an extensible picture processing suite for electron microscopy. J Struct Biol. 2007;157:38–46. pmid:16859925
  73. 73.
    Kremer JR, Mastronarde DN, McIntosh JR. Laptop visualization of three-dimensional picture information utilizing IMOD. J Struct Biol. 1996;116:71–76. pmid:8742726
RELATED ARTICLES

LEAVE A REPLY

Please enter your comment!
Please enter your name here

Most Popular

Recent Comments