Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell ; 187(9): 2158-2174.e19, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38604175

ABSTRACT

Centriole biogenesis, as in most organelle assemblies, involves the sequential recruitment of sub-structural elements that will support its function. To uncover this process, we correlated the spatial location of 24 centriolar proteins with structural features using expansion microscopy. A time-series reconstruction of protein distributions throughout human procentriole assembly unveiled the molecular architecture of the centriole biogenesis steps. We found that the process initiates with the formation of a naked cartwheel devoid of microtubules. Next, the bloom phase progresses with microtubule blade assembly, concomitantly with radial separation and rapid cartwheel growth. In the subsequent elongation phase, the tubulin backbone grows linearly with the recruitment of the A-C linker, followed by proteins of the inner scaffold (IS). By following six structural modules, we modeled 4D assembly of the human centriole. Collectively, this work provides a framework to investigate the spatial and temporal assembly of large macromolecules.


Subject(s)
Centrioles , Microtubules , Centrioles/metabolism , Humans , Microtubules/metabolism , Tubulin/metabolism , Cell Cycle Proteins/metabolism
2.
EMBO J ; 41(21): e112107, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36125182

ABSTRACT

Over the course of evolution, the centrosome function has been conserved in most eukaryotes, but its core architecture has evolved differently in some clades, with the presence of centrioles in humans and a spindle pole body (SPB) in yeast. Similarly, the composition of these two core elements has diverged, with the exception of Centrin and SFI1, which form a complex in yeast to initiate SPB duplication. However, it remains unclear whether this complex exists at centrioles and whether its function has been conserved. Here, using expansion microscopy, we demonstrate that human SFI1 is a centriolar protein that associates with a pool of Centrin at the distal end of the centriole. We also find that both proteins are recruited early during procentriole assembly and that depletion of SFI1 results in the loss of the distal pool of Centrin, without altering centriole duplication. Instead, we show that SFI1/Centrin complex is essential for centriolar architecture, CEP164 distribution, and CP110 removal during ciliogenesis. Together, our work reveals a conserved SFI1/Centrin module displaying divergent functions between mammals and yeast.


Subject(s)
Calcium-Binding Proteins , Cell Cycle Proteins , Centrioles , Animals , Humans , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spindle Pole Bodies/metabolism , Calcium-Binding Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 120(9): e2102569120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36802443

ABSTRACT

In the human genome, about 750 genes contain one intron excised by the minor spliceosome. This spliceosome comprises its own set of snRNAs, among which U4atac. Its noncoding gene, RNU4ATAC, has been found mutated in Taybi-Linder (TALS/microcephalic osteodysplastic primordial dwarfism type 1), Roifman (RFMN), and Lowry-Wood (LWS) syndromes. These rare developmental disorders, whose physiopathological mechanisms remain unsolved, associate ante- and post-natal growth retardation, microcephaly, skeletal dysplasia, intellectual disability, retinal dystrophy, and immunodeficiency. Here, we report bi-allelic RNU4ATAC mutations in five patients presenting with traits suggestive of the Joubert syndrome (JBTS), a well-characterized ciliopathy. These patients also present with traits typical of TALS/RFMN/LWS, thus widening the clinical spectrum of RNU4ATAC-associated disorders and indicating ciliary dysfunction as a mechanism downstream of minor splicing defects. Intriguingly, all five patients carry the n.16G>A mutation, in the Stem II domain, either at the homozygous or compound heterozygous state. A gene ontology term enrichment analysis on minor intron-containing genes reveals that the cilium assembly process is over-represented, with no less than 86 cilium-related genes containing at least one minor intron, among which there are 23 ciliopathy-related genes. The link between RNU4ATAC mutations and ciliopathy traits is supported by alterations of primary cilium function in TALS and JBTS-like patient fibroblasts, as well as by u4atac zebrafish model, which exhibits ciliopathy-related phenotypes and ciliary defects. These phenotypes could be rescued by WT but not by pathogenic variants-carrying human U4atac. Altogether, our data indicate that alteration of cilium biogenesis is part of the physiopathological mechanisms of TALS/RFMN/LWS, secondarily to defects of minor intron splicing.


Subject(s)
Ciliopathies , Spliceosomes , Female , Animals , Humans , Spliceosomes/genetics , RNA, Small Nuclear/genetics , Zebrafish/genetics , Fetal Growth Retardation/genetics , Mutation , Ciliopathies/genetics
4.
Semin Cell Dev Biol ; 137: 16-25, 2023 03 15.
Article in English | MEDLINE | ID: mdl-34896019

ABSTRACT

Centrioles are microtubule-based cell organelles present in most eukaryotes. They participate in the control of cell division as part of the centrosome, the major microtubule-organizing center of the cell, and are also essential for the formation of primary and motile cilia. During centriole assembly as well as across its lifetime, centriolar tubulin display marks defined by post-translational modifications (PTMs), such as glutamylation or acetylation. To date, the functions of these PTMs at centrioles are not well understood, although pioneering experiments suggest a role in the stability of this organelle. Here, we review the current knowledge regarding PTMs at centrioles with a particular focus on a possible link between these modifications and centriole's architecture, and propose possible hypothesis regarding centriolar tubulin PTMs's function.


Subject(s)
Centrioles , Tubulin , Tubulin/genetics , Microtubule-Organizing Center , Microtubules , Cilia
5.
Nat Methods ; 19(2): 216-222, 2022 02.
Article in English | MEDLINE | ID: mdl-35027766

ABSTRACT

Cryofixation has proven to be the gold standard for efficient preservation of native cell ultrastructure compared to chemical fixation, but this approach is not widely used in fluorescence microscopy owing to implementation challenges. Here, we develop Cryo-ExM, a method that preserves native cellular organization by coupling cryofixation with expansion microscopy. This method bypasses artifacts associated with chemical fixation and its simplicity will contribute to its widespread use in super-resolution microscopy.


Subject(s)
Cryopreservation/methods , Microscopy, Fluorescence/methods , Animals , Cell Line , Chlamydomonas reinhardtii/cytology , Cryopreservation/instrumentation , Cytoskeleton , Epitopes , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice
6.
PLoS Biol ; 20(6): e3001649, 2022 06.
Article in English | MEDLINE | ID: mdl-35709082

ABSTRACT

Inherited retinal degeneration due to loss of photoreceptor cells is a leading cause of human blindness. These cells possess a photosensitive outer segment linked to the cell body through the connecting cilium (CC). While structural defects of the CC have been associated with retinal degeneration, its nanoscale molecular composition, assembly, and function are barely known. Here, using expansion microscopy and electron microscopy, we reveal the molecular architecture of the CC and demonstrate that microtubules are linked together by a CC inner scaffold containing POC5, CENTRIN, and FAM161A. Dissecting CC inner scaffold assembly during photoreceptor development in mouse revealed that it acts as a structural zipper, progressively bridging microtubule doublets and straightening the CC. Furthermore, we show that Fam161a disruption in mouse leads to specific CC inner scaffold loss and triggers microtubule doublet spreading, prior to outer segment collapse and photoreceptor degeneration, suggesting a molecular mechanism for a subtype of retinitis pigmentosa.


Subject(s)
Retinal Degeneration , Retinitis Pigmentosa , Animals , Cilia , Eye Proteins , Mice , Microtubules
7.
PLoS Biol ; 20(9): e3001782, 2022 09.
Article in English | MEDLINE | ID: mdl-36070319

ABSTRACT

In metazoa, cilia assembly is a cellular process that starts with centriole to basal body maturation, migration to the cell surface, and docking to the plasma membrane. Basal body docking involves the interaction of both the distal end of the basal body and the transition fibers/distal appendages, with the plasma membrane. Mutations in numerous genes involved in basal body docking and transition zone assembly are associated with the most severe ciliopathies, highlighting the importance of these events in cilium biogenesis. In this context, the ciliate Paramecium has been widely used as a model system to study basal body and cilia assembly. However, despite the evolutionary conservation of cilia assembly events across phyla, whether the same molecular players are functionally conserved, is not fully known. Here, we demonstrated that CEP90, FOPNL, and OFD1 are evolutionary conserved proteins crucial for ciliogenesis. Using ultrastructure expansion microscopy, we unveiled that these proteins localize at the distal end of both centrioles/basal bodies in Paramecium and mammalian cells. Moreover, we found that these proteins are recruited early during centriole duplication on the external surface of the procentriole. Functional analysis performed both in Paramecium and mammalian cells demonstrate the requirement of these proteins for distal appendage assembly and basal body docking. Finally, we show that mammalian centrioles require another component, Moonraker (MNR), to recruit OFD1, FOPNL, and CEP90, which will then recruit the distal appendage proteins CEP83, CEP89, and CEP164. Altogether, we propose that this OFD1, FOPNL, and CEP90 functional module is required to determine in mammalian cells the future position of distal appendage proteins.


Subject(s)
Centrioles/metabolism , Cilia/ultrastructure , Paramecium/metabolism , Animals , Cell Membrane , Centrioles/chemistry , Cilia/metabolism , Mammals , Paramecium/chemistry , Paramecium/cytology
8.
EMBO J ; 39(22): e106246, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32954513

ABSTRACT

Centrioles are evolutionarily conserved barrels of microtubule triplets that form the core of the centrosome and the base of the cilium. While the crucial role of the proximal region in centriole biogenesis has been well documented, its native architecture and evolutionary conservation remain relatively unexplored. Here, using cryo-electron tomography of centrioles from four evolutionarily distant species, we report on the architectural diversity of the centriole's proximal cartwheel-bearing region. Our work reveals that the cartwheel central hub is constructed from a stack of paired rings with cartwheel inner densities inside. In both Paramecium and Chlamydomonas, the repeating structural unit of the cartwheel has a periodicity of 25 nm and consists of three ring pairs, with 6 radial spokes emanating and merging into a single bundle that connects to the microtubule triplet via the D2-rod and the pinhead. Finally, we identified that the cartwheel is indirectly connected to the A-C linker through the triplet base structure extending from the pinhead. Together, our work provides unprecedented evolutionary insights into the architecture of the centriole proximal region, which underlies centriole biogenesis.


Subject(s)
Centrioles/physiology , Centrioles/ultrastructure , Electron Microscope Tomography/methods , Centrosome , Chlamydomonas reinhardtii/physiology , Cilia , Humans , Microtubules , Models, Molecular , Naegleria/physiology , Paramecium tetraurelia/physiology
9.
J Cell Sci ; 135(24)2022 12 15.
Article in English | MEDLINE | ID: mdl-36524422

ABSTRACT

The budding and fission yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe have served as invaluable model organisms to study conserved fundamental cellular processes. Although super-resolution microscopy has in recent years paved the way to a better understanding of the spatial organization of molecules in cells, its wide use in yeasts has remained limited due to the specific know-how and instrumentation required, contrasted with the relative ease of endogenous tagging and live-cell fluorescence microscopy. To facilitate super-resolution microscopy in yeasts, we have extended the ultrastructure expansion microscopy (U-ExM) method to both S. cerevisiae and S. pombe, enabling a 4-fold isotropic expansion. We demonstrate that U-ExM allows imaging of the microtubule cytoskeleton and its associated spindle pole body, notably unveiling the Sfi1p-Cdc31p spatial organization on the appendage bridge structure. In S. pombe, we validate the method by monitoring the homeostatic regulation of nuclear pore complex number through the cell cycle. Combined with NHS-ester pan-labelling, which provides a global cellular context, U-ExM reveals the subcellular organization of these two yeast models and provides a powerful new method to augment the already extensive yeast toolbox. This article has an associated First Person interview with Kerstin Hinterndorfer and Felix Mikus, two of the joint first authors of the paper.


Subject(s)
Saccharomyces cerevisiae Proteins , Schizosaccharomyces , Humans , Schizosaccharomyces/metabolism , Saccharomyces cerevisiae/metabolism , Microscopy , Saccharomyces cerevisiae Proteins/metabolism , Spindle Pole Bodies/metabolism
11.
PLoS Biol ; 19(3): e3001020, 2021 03.
Article in English | MEDLINE | ID: mdl-33705377

ABSTRACT

Malaria is caused by unicellular Plasmodium parasites. Plasmodium relies on diverse microtubule cytoskeletal structures for its reproduction, multiplication, and dissemination. Due to the small size of this parasite, its cytoskeleton has been primarily observable by electron microscopy (EM). Here, we demonstrate that the nanoscale cytoskeleton organisation is within reach using ultrastructure expansion microscopy (U-ExM). In developing microgametocytes, U-ExM allows monitoring the dynamic assembly of axonemes and concomitant tubulin polyglutamylation in whole cells. In the invasive merozoite and ookinete forms, U-ExM unveils the diversity across Plasmodium stages and species of the subpellicular microtubule arrays that confer cell rigidity. In ookinetes, we additionally identify an apical tubulin ring (ATR) that colocalises with markers of the conoid in related apicomplexan parasites. This tubulin-containing structure was presumed to be lost in Plasmodium despite its crucial role in motility and invasion in other apicomplexans. Here, U-ExM reveals that a divergent and considerably reduced form of the conoid is actually conserved in Plasmodium species.


Subject(s)
Cytoskeleton/ultrastructure , Microtubules/ultrastructure , Toxoplasma/ultrastructure , Animals , Cytoskeleton/metabolism , Malaria/metabolism , Malaria/parasitology , Microscopy, Electron/methods , Microtubules/metabolism , Parasites , Plasmodium/metabolism , Plasmodium/pathogenicity , Plasmodium/ultrastructure , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Tubulin
12.
Nat Methods ; 17(7): 726-733, 2020 07.
Article in English | MEDLINE | ID: mdl-32572233

ABSTRACT

Super-resolution microscopies have become an established tool in biological research. However, imaging throughput remains a main bottleneck in acquiring large datasets required for quantitative biology. Here we describe multifocal flat illumination for field-independent imaging (mfFIFI). By integrating mfFIFI into an instant structured illumination microscope (iSIM), we extend the field of view (FOV) to >100 × 100 µm2 while maintaining high-speed, multicolor, volumetric imaging at double the diffraction-limited resolution. We further extend the effective FOV by stitching adjacent images for fast live-cell super-resolution imaging of dozens of cells. Finally, we combine our flat-fielded iSIM with ultrastructure expansion microscopy to collect three-dimensional (3D) images of hundreds of centrioles in human cells, or thousands of purified Chlamydomonas reinhardtii centrioles, per hour at an effective resolution of ~35 nm. Classification and particle averaging of these large datasets enables 3D mapping of posttranslational modifications of centriolar microtubules, revealing differences in their coverage and positioning.


Subject(s)
Microscopy, Fluorescence/methods , Animals , COS Cells , Centrioles/ultrastructure , Chlorocebus aethiops , Humans , Imaging, Three-Dimensional , Lighting , Microtubules/ultrastructure
13.
Nat Methods ; 16(1): 71-74, 2019 01.
Article in English | MEDLINE | ID: mdl-30559430

ABSTRACT

Determining the structure and composition of macromolecular assemblies is a major challenge in biology. Here we describe ultrastructure expansion microscopy (U-ExM), an extension of expansion microscopy that allows the visualization of preserved ultrastructures by optical microscopy. This method allows for near-native expansion of diverse structures in vitro and in cells; when combined with super-resolution microscopy, it unveiled details of ultrastructural organization, such as centriolar chirality, that could otherwise be observed only by electron microscopy.


Subject(s)
Microscopy, Electron/methods , Microscopy, Fluorescence/methods , Microtubules/metabolism , Stereoisomerism
14.
Bioessays ; 40(4): e1700241, 2018 04.
Article in English | MEDLINE | ID: mdl-29508910

ABSTRACT

The cartwheel is a striking structure critical for building the centriole, a microtubule-based organelle fundamental for organizing centrosomes, cilia, and flagella. Over the last 50 years, the cartwheel has been described in many systems using electron microscopy, but the molecular nature of its constituent building blocks and their assembly mechanisms have long remained mysterious. Here, we review discoveries that led to the current understanding of cartwheel structure, assembly, and function. We focus on the key role of SAS-6 protein self-organization, both for building the signature ring-like structure with hub and spokes, as well as for their vertical stacking. The resemblance of assembly intermediates in vitro and in vivo leads us to propose a novel registration step in cartwheel biogenesis, whereby stacked SAS-6-containing rings are put in register through interactions with peripheral elements anchored to microtubules. We conclude by evoking some avenues for further uncovering cartwheel and centriole assembly mechanisms.


Subject(s)
Centrioles/metabolism , Organelles/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/genetics , Centrioles/ultrastructure , Cilia/genetics , Cilia/metabolism , Cilia/ultrastructure , Microscopy, Electron , Microtubules/genetics , Microtubules/metabolism , Microtubules/ultrastructure , Organelles/genetics , Organelles/ultrastructure
15.
Curr Opin Cell Biol ; 88: 102361, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38648677

ABSTRACT

Cilia and flagella are specialized eukaryotic organelles projecting from the surface of eukaryotic cells that play a central role in various physiological processes, including cell motility, sensory perception, and signal transduction. At the base of these structures lies the ciliary transition zone, a pivotal region that functions as a gatekeeper and communication hub for ciliary activities. Despite its crucial role, the intricacies of its architecture remain poorly understood, especially given the variations in its organization across different cell types and species. In this review, we explore the molecular architecture of the ciliary transition zone, with a particular focus on recent findings obtained using cryotomography and super-resolution imaging techniques.


Subject(s)
Cilia , Cilia/metabolism , Cilia/chemistry , Animals , Humans , Flagella/metabolism
16.
MicroPubl Biol ; 20242024.
Article in English | MEDLINE | ID: mdl-38774216

ABSTRACT

Visualization of organelles using expansion microscopy has been previously applied to Caenorhadbitis elegans adult gonads or worms. However, its application to embryos has remained a challenge due to the protective eggshell barrier. Here, by combining freeze-cracking and ultrastructure expansion microscopy (U-ExM), we demonstrate a four-time isotropic expansion of C. elegans embryos. As an example structure, we chose the nuclear pore and demonstrate that we achieve sufficient resolution to distinguish them individually. Our work provides proof of principle for U-ExM in C. elegans embryos, which will be applicable for imaging a wide range of cellular structures in this model system.

17.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38617270

ABSTRACT

Centrosomes have critical roles in microtubule organization and in cell signaling.1-8 However, the mechanisms that regulate centrosome function are not fully defined, and thus how defects in centrosomal regulation contribute to disease is incompletely understood. From functional genomic analyses, we find here that PPP2R3C, a PP2A phosphatase subunit, is a distal centriole protein and functional partner of centriolar proteins CEP350 and FOP. We further show that a key function of PPP2R3C is to counteract the kinase activity of MAP3K1. In support of this model, MAP3K1 knockout suppresses growth defects caused by PPP2R3C inactivation, and MAP3K1 and PPP2R3C have opposing effects on basal and microtubule stress-induced JNK signaling. Illustrating the importance of balanced MAP3K1 and PPP2R3C activities, acute overexpression of MAP3K1 severely inhibits centrosome function and triggers rapid centriole disintegration. Additionally, inactivating PPP2R3C mutations and activating MAP3K1 mutations both cause congenital syndromes characterized by gonadal dysgenesis.9-15 As a syndromic PPP2R3C variant is defective in centriolar localization and binding to centriolar protein FOP, we propose that imbalanced activity of this centrosomal kinase-phosphatase pair is the shared cause of these disorders. Thus, our findings reveal a new centrosomal phospho-regulatory module, shed light on disorders of gonadal development, and illustrate the power of systems genetics to identify previously unrecognized gene functions.

18.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504136

ABSTRACT

For 15 years, gene therapy has been viewed as a beacon of hope for inherited retinal diseases. Many preclinical investigations have centered around vectors with maximal gene expression capabilities, yet despite efficient gene transfer, minimal physiological improvements have been observed in various ciliopathies. Retinitis pigmentosa-type 28 (RP28) is the consequence of bi-allelic null mutations in the FAM161A, an essential protein for the structure of the photoreceptor connecting cilium (CC). In its absence, cilia become disorganized, leading to outer segment collapses and vision impairment. Within the human retina, FAM161A has two isoforms: the long one with exon 4, and the short one without it. To restore CC in Fam161a-deficient mice shortly after the onset of cilium disorganization, we compared AAV vectors with varying promoter activities, doses, and human isoforms. While all vectors improved cell survival, only the combination of both isoforms using the weak FCBR1-F0.4 promoter enabled precise FAM161A expression in the CC and enhanced retinal function. Our investigation into FAM161A gene replacement for RP28 emphasizes the importance of precise therapeutic gene regulation, appropriate vector dosing, and delivery of both isoforms. This precision is pivotal for secure gene therapy involving structural proteins like FAM161A.


Subject(s)
Retinitis Pigmentosa , Animals , Mice , Humans , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Exons , Protein Isoforms/genetics , Protein Isoforms/metabolism , Genetic Therapy , Eye Proteins/genetics , Eye Proteins/chemistry , Eye Proteins/metabolism
19.
Elife ; 122024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441556

ABSTRACT

From a cohort of 167 infertile patients suffering from multiple morphological abnormalities of the flagellum (MMAF), pathogenic bi-allelic mutations were identified in the CCDC146 gene. In somatic cells, CCDC146 is located at the centrosome and at multiple microtubule-related organelles during mitotic division, suggesting that it is a microtubule-associated protein (MAP). To decipher the molecular pathogenesis of infertility associated with CCDC146 mutations, a Ccdc146 knock-out (KO) mouse line was created. KO male mice were infertile, and sperm exhibited a phenotype identical to CCDC146 mutated patients. CCDC146 expression starts during late spermiogenesis. In the spermatozoon, the protein is conserved but is not localized to centrioles, unlike in somatic cells, rather it is present in the axoneme at the level of microtubule doublets. Expansion microscopy associated with the use of the detergent sarkosyl to solubilize microtubule doublets suggests that the protein may be a microtubule inner protein (MIP). At the subcellular level, the absence of CCDC146 impacted all microtubule-based organelles such as the manchette, the head-tail coupling apparatus (HTCA), and the axoneme. Through this study, a new genetic cause of infertility and a new factor in the formation and/or structure of the sperm axoneme were characterized.


Subject(s)
Abnormalities, Multiple , Infertility, Male , Animals , Humans , Male , Mice , Centrioles , Infertility, Male/genetics , Mice, Knockout , Microtubule-Associated Proteins/genetics , Semen
20.
Med Sci (Paris) ; 39(4): 351-358, 2023 Apr.
Article in French | MEDLINE | ID: mdl-37094268

ABSTRACT

Most cellular imaging techniques, such as light or electron microscopy, require that the biological sample is first fixed by chemical cross-linking agents. This necessary step is also known to damage molecular nanostructures or even sub-cellular organization. To overcome this problem, another fixation approach was invented more than 40 years ago, which consists in cryo-fixing biological samples, thus allowing to preserve their native state. However, this method has been scarcely used in light microscopy due to the complexity of its implementation. In this review, we present a recently developed super-resolution method called expansion microscopy, which, when coupled with cryo-fixation, allows to visualize at a nanometric resolution the cell architecture as close as possible to its native state.


Title: L'organisation native de la cellule révélée grâce à la cryo-microscopie à expansion. Abstract: La plupart des techniques d'imagerie cellulaire, telles que la microscopie photonique ou la microscopie électronique, nécessitent que l'échantillon biologique soit préalablement fixé par des agents chimiques, une étape qui est connue pour endommager l'organisation sub-cellulaire. Pour pallier à ce problème, la cryo-fixation, inventée il y a plus de 40 ans, consiste à vitrifier les échantillons biologiques afin de préserver leur état natif. Cette méthode n'avait cependant été que très peu utilisée en microscopie photonique. Dans cette revue, nous présentons en détail la microscopie d'expansion, une technique de super-résolution développée récemment et qui, couplée à la cryo-fixation, permet de visualiser l'architecture cellulaire au plus près de son état natif.


Subject(s)
Cryopreservation , Humans , Microscopy, Electron , Cryopreservation/methods , Cryoelectron Microscopy/methods
SELECTION OF CITATIONS
SEARCH DETAIL