Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 253
Filter
1.
Cell ; 184(9): 2412-2429.e16, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33852913

ABSTRACT

Cellular versatility depends on accurate trafficking of diverse proteins to their organellar destinations. For the secretory pathway (followed by approximately 30% of all proteins), the physical nature of the vessel conducting the first portage (endoplasmic reticulum [ER] to Golgi apparatus) is unclear. We provide a dynamic 3D view of early secretory compartments in mammalian cells with isotropic resolution and precise protein localization using whole-cell, focused ion beam scanning electron microscopy with cryo-structured illumination microscopy and live-cell synchronized cargo release approaches. Rather than vesicles alone, the ER spawns an elaborate, interwoven tubular network of contiguous lipid bilayers (ER exit site) for protein export. This receptacle is capable of extending microns along microtubules while still connected to the ER by a thin neck. COPII localizes to this neck region and dynamically regulates cargo entry from the ER, while COPI acts more distally, escorting the detached, accelerating tubular entity on its way to joining the Golgi apparatus through microtubule-directed movement.


Subject(s)
COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Microtubules/metabolism , Ubiquitin-Protein Ligases/metabolism , Biological Transport, Active , HeLa Cells , Humans , Protein Transport
2.
Cell ; 177(6): 1522-1535.e14, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31130380

ABSTRACT

Metabolic coordination between neurons and astrocytes is critical for the health of the brain. However, neuron-astrocyte coupling of lipid metabolism, particularly in response to neural activity, remains largely uncharacterized. Here, we demonstrate that toxic fatty acids (FAs) produced in hyperactive neurons are transferred to astrocytic lipid droplets by ApoE-positive lipid particles. Astrocytes consume the FAs stored in lipid droplets via mitochondrial ß-oxidation in response to neuronal activity and turn on a detoxification gene expression program. Our findings reveal that FA metabolism is coupled in neurons and astrocytes to protect neurons from FA toxicity during periods of enhanced activity. This coordinated mechanism for metabolizing FAs could underlie both homeostasis and a variety of disease states of the brain.


Subject(s)
Astrocytes/metabolism , Fatty Acids/metabolism , Neurons/metabolism , Animals , Apolipoproteins E/metabolism , Apolipoproteins E/physiology , Astrocytes/physiology , Brain/metabolism , Fatty Acids/toxicity , Homeostasis , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
3.
Cell ; 179(1): 147-164.e20, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31539493

ABSTRACT

Long-distance RNA transport enables local protein synthesis at metabolically-active sites distant from the nucleus. This process ensures an appropriate spatial organization of proteins, vital to polarized cells such as neurons. Here, we present a mechanism for RNA transport in which RNA granules "hitchhike" on moving lysosomes. In vitro biophysical modeling, live-cell microscopy, and unbiased proximity labeling proteomics reveal that annexin A11 (ANXA11), an RNA granule-associated phosphoinositide-binding protein, acts as a molecular tether between RNA granules and lysosomes. ANXA11 possesses an N-terminal low complexity domain, facilitating its phase separation into membraneless RNA granules, and a C-terminal membrane binding domain, enabling interactions with lysosomes. RNA granule transport requires ANXA11, and amyotrophic lateral sclerosis (ALS)-associated mutations in ANXA11 impair RNA granule transport by disrupting their interactions with lysosomes. Thus, ANXA11 mediates neuronal RNA transport by tethering RNA granules to actively-transported lysosomes, performing a critical cellular function that is disrupted in ALS.


Subject(s)
Annexins/metabolism , Axonal Transport/physiology , Cytoplasmic Granules/metabolism , Lysosomes/metabolism , RNA/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Genetically Modified , Annexins/genetics , Axons/metabolism , Cell Line, Tumor , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mutation , Protein Binding , Rats/embryology , Rats, Sprague-Dawley , Transfection , Zebrafish
4.
Cell ; 175(5): 1430-1442.e17, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30454650

ABSTRACT

In eukaryotic cells, organelles and the cytoskeleton undergo highly dynamic yet organized interactions capable of orchestrating complex cellular functions. Visualizing these interactions requires noninvasive, long-duration imaging of the intracellular environment at high spatiotemporal resolution and low background. To achieve these normally opposing goals, we developed grazing incidence structured illumination microscopy (GI-SIM) that is capable of imaging dynamic events near the basal cell cortex at 97-nm resolution and 266 frames/s over thousands of time points. We employed multi-color GI-SIM to characterize the fast dynamic interactions of diverse organelles and the cytoskeleton, shedding new light on the complex behaviors of these structures. Precise measurements of microtubule growth or shrinkage events helped distinguish among models of microtubule dynamic instability. Analysis of endoplasmic reticulum (ER) interactions with other organelles or microtubules uncovered new ER remodeling mechanisms, such as hitchhiking of the ER on motile organelles. Finally, ER-mitochondria contact sites were found to promote both mitochondrial fission and fusion.


Subject(s)
Endoplasmic Reticulum/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Microscopy, Fluorescence
5.
Nature ; 626(7997): 169-176, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38267577

ABSTRACT

To coordinate cellular physiology, eukaryotic cells rely on the rapid exchange of molecules at specialized organelle-organelle contact sites1,2. Endoplasmic reticulum-mitochondrial contact sites (ERMCSs) are particularly vital communication hubs, playing key roles in the exchange of signalling molecules, lipids and metabolites3,4. ERMCSs are maintained by interactions between complementary tethering molecules on the surface of each organelle5,6. However, due to the extreme sensitivity of these membrane interfaces to experimental perturbation7,8, a clear understanding of their nanoscale organization and regulation is still lacking. Here we combine three-dimensional electron microscopy with high-speed molecular tracking of a model organelle tether, Vesicle-associated membrane protein (VAMP)-associated protein B (VAPB), to map the structure and diffusion landscape of ERMCSs. We uncovered dynamic subdomains within VAPB contact sites that correlate with ER membrane curvature and undergo rapid remodelling. We show that VAPB molecules enter and leave ERMCSs within seconds, despite the contact site itself remaining stable over much longer time scales. This metastability allows ERMCSs to remodel with changes in the physiological environment to accommodate metabolic needs of the cell. An amyotrophic lateral sclerosis-associated mutation in VAPB perturbs these subdomains, likely impairing their remodelling capacity and resulting in impaired interorganelle communication. These results establish high-speed single-molecule imaging as a new tool for mapping the structure of contact site interfaces and reveal that the diffusion landscape of VAPB at contact sites is a crucial component of ERMCS homeostasis.


Subject(s)
Endoplasmic Reticulum , Mitochondria , Mitochondrial Membranes , Movement , Vesicular Transport Proteins , Humans , Amyotrophic Lateral Sclerosis/genetics , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/ultrastructure , Microscopy, Electron , Imaging, Three-Dimensional , Binding Sites , Diffusion , Time Factors , Mutation , Homeostasis
6.
Cell ; 158(3): 522-33, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083867

ABSTRACT

Proteins destined for the cell surface are first assessed in the endoplasmic reticulum (ER) for proper folding before release into the secretory pathway. This ensures that defective proteins are normally prevented from entering the extracellular environment, where they could be disruptive. Here, we report that, when ER folding capacity is saturated during stress, misfolded glycosylphosphatidylinositol-anchored proteins dissociate from resident ER chaperones, engage export receptors, and quantitatively leave the ER via vesicular transport to the Golgi. Clearance from the ER commences within minutes of acute ER stress, before the transcriptional component of the unfolded protein response is activated. These aberrant proteins then access the cell surface transiently before destruction in lysosomes. Inhibiting this stress-induced pathway by depleting the ER-export receptors leads to aggregation of the ER-retained misfolded protein. Thus, this rapid response alleviates the elevated burden of misfolded proteins in the ER at the onset of ER stress, promoting protein homeostasis in the ER.


Subject(s)
Endoplasmic Reticulum Stress , Lysosomes/metabolism , Secretory Pathway , Animals , Cell Line , Humans , Mice , Prions/metabolism , Protein Folding , Rats , Unfolded Protein Response
7.
Cell ; 158(4): 822-832, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126787

ABSTRACT

Molecular motors in cells typically produce highly directed motion; however, the aggregate, incoherent effect of all active processes also creates randomly fluctuating forces, which drive diffusive-like, nonthermal motion. Here, we introduce force-spectrum-microscopy (FSM) to directly quantify random forces within the cytoplasm of cells and thereby probe stochastic motor activity. This technique combines measurements of the random motion of probe particles with independent micromechanical measurements of the cytoplasm to quantify the spectrum of force fluctuations. Using FSM, we show that force fluctuations substantially enhance intracellular movement of small and large components. The fluctuations are three times larger in malignant cells than in their benign counterparts. We further demonstrate that vimentin acts globally to anchor organelles against randomly fluctuating forces in the cytoplasm, with no effect on their magnitude. Thus, FSM has broad applications for understanding the cytoplasm and its intracellular processes in relation to cell physiology in healthy and diseased states.


Subject(s)
Cytoplasm/chemistry , Microscopy, Atomic Force/methods , Animals , Biomechanical Phenomena , Embryo, Mammalian/cytology , Fibroblasts/chemistry , Mice , Proteins/chemistry , Vimentin/chemistry
8.
Cell ; 155(6): 1220-31, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24315094

ABSTRACT

The final cleavage event that terminates cell division, abscission of the small, dense intercellular bridge, has been particularly challenging to resolve. Here, we describe imaging innovations that helped answer long-standing questions about the mechanism of abscission. We further explain how computational modeling of high-resolution data was employed to test hypotheses and generate additional insights. We present the model that emerges from application of these complimentary approaches. Similar experimental strategies will undoubtedly reveal exciting details about other underresolved cellular structures.


Subject(s)
Cytokinesis , Models, Biological , Plant Cells/ultrastructure , Animals , Cell Physiological Phenomena , Humans , Plant Cells/metabolism
9.
Nature ; 601(7891): 132-138, 2022 01.
Article in English | MEDLINE | ID: mdl-34912111

ABSTRACT

Organelles move along differentially modified microtubules to establish and maintain their proper distributions and functions1,2. However, how cells interpret these post-translational microtubule modification codes to selectively regulate organelle positioning remains largely unknown. The endoplasmic reticulum (ER) is an interconnected network of diverse morphologies that extends promiscuously throughout the cytoplasm3, forming abundant contacts with other organelles4. Dysregulation of endoplasmic reticulum morphology is tightly linked to neurologic disorders and cancer5,6. Here we demonstrate that three membrane-bound endoplasmic reticulum proteins preferentially interact with different microtubule populations, with CLIMP63 binding centrosome microtubules, kinectin (KTN1) binding perinuclear polyglutamylated microtubules, and p180 binding glutamylated microtubules. Knockout of these proteins or manipulation of microtubule populations and glutamylation status results in marked changes in endoplasmic reticulum positioning, leading to similar redistributions of other organelles. During nutrient starvation, cells modulate CLIMP63 protein levels and p180-microtubule binding to bidirectionally move endoplasmic reticulum and lysosomes for proper autophagic responses.


Subject(s)
Centrosome/metabolism , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Tubulin/metabolism , Animals , Autophagy , Biological Transport , Cell Line , Glutamic Acid/metabolism , Humans , Membrane Proteins/metabolism , Microtubules/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
10.
EMBO J ; 42(6): e112863, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36807601

ABSTRACT

The Hippo pathway was originally discovered to control tissue growth in Drosophila and includes the Hippo kinase (Hpo; MST1/2 in mammals), scaffold protein Salvador (Sav; SAV1 in mammals) and the Warts kinase (Wts; LATS1/2 in mammals). The Hpo kinase is activated by binding to Crumbs-Expanded (Crb-Ex) and/or Merlin-Kibra (Mer-Kib) proteins at the apical domain of epithelial cells. Here we show that activation of Hpo also involves the formation of supramolecular complexes with properties of a biomolecular condensate, including concentration dependence and sensitivity to starvation, macromolecular crowding, or 1,6-hexanediol treatment. Overexpressing Ex or Kib induces formation of micron-scale Hpo condensates in the cytoplasm, rather than at the apical membrane. Several Hippo pathway components contain unstructured low-complexity domains and purified Hpo-Sav complexes undergo phase separation in vitro. Formation of Hpo condensates is conserved in human cells. We propose that apical Hpo kinase activation occurs in phase separated "signalosomes" induced by clustering of upstream pathway components.


Subject(s)
Drosophila Proteins , Hippo Signaling Pathway , Animals , Humans , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Neurofibromin 2/metabolism , Drosophila melanogaster/metabolism , Mammals , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
11.
EMBO J ; 42(24): e114054, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37933600

ABSTRACT

Cristae are high-curvature structures in the inner mitochondrial membrane (IMM) that are crucial for ATP production. While cristae-shaping proteins have been defined, analogous lipid-based mechanisms have yet to be elucidated. Here, we combine experimental lipidome dissection with multi-scale modeling to investigate how lipid interactions dictate IMM morphology and ATP generation. When modulating phospholipid (PL) saturation in engineered yeast strains, we observed a surprisingly abrupt breakpoint in IMM topology driven by a continuous loss of ATP synthase organization at cristae ridges. We found that cardiolipin (CL) specifically buffers the inner mitochondrial membrane against curvature loss, an effect that is independent of ATP synthase dimerization. To explain this interaction, we developed a continuum model for cristae tubule formation that integrates both lipid and protein-mediated curvatures. This model highlighted a snapthrough instability, which drives IMM collapse upon small changes in membrane properties. We also showed that cardiolipin is essential in low-oxygen conditions that promote PL saturation. These results demonstrate that the mechanical function of cardiolipin is dependent on the surrounding lipid and protein components of the IMM.


Subject(s)
Cardiolipins , Lipidomics , Cardiolipins/metabolism , Mitochondrial Membranes/metabolism , Phospholipids/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism
12.
Nature ; 599(7883): 141-146, 2021 11.
Article in English | MEDLINE | ID: mdl-34616042

ABSTRACT

Cells contain hundreds of organelles and macromolecular assemblies. Obtaining a complete understanding of their intricate organization requires the nanometre-level, three-dimensional reconstruction of whole cells, which is only feasible with robust and scalable automatic methods. Here, to support the development of such methods, we annotated up to 35 different cellular organelle classes-ranging from endoplasmic reticulum to microtubules to ribosomes-in diverse sample volumes from multiple cell types imaged at a near-isotropic resolution of 4 nm per voxel with focused ion beam scanning electron microscopy (FIB-SEM)1. We trained deep learning architectures to segment these structures in 4 nm and 8 nm per voxel FIB-SEM volumes, validated their performance and showed that automatic reconstructions can be used to directly quantify previously inaccessible metrics including spatial interactions between cellular components. We also show that such reconstructions can be used to automatically register light and electron microscopy images for correlative studies. We have created an open data and open-source web repository, 'OpenOrganelle', to share the data, computer code and trained models, which will enable scientists everywhere to query and further improve automatic reconstruction of these datasets.


Subject(s)
Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Organelles/ultrastructure , Animals , Biomarkers/analysis , COS Cells , Cell Size , Chlorocebus aethiops , Datasets as Topic , Deep Learning , Endoplasmic Reticulum , HeLa Cells , Humans , Information Dissemination , Microscopy, Fluorescence , Microtubules , Reproducibility of Results , Ribosomes
13.
Nature ; 599(7883): 147-151, 2021 11.
Article in English | MEDLINE | ID: mdl-34616045

ABSTRACT

Understanding cellular architecture is essential for understanding biology. Electron microscopy (EM) uniquely visualizes cellular structures with nanometre resolution. However, traditional methods, such as thin-section EM or EM tomography, have limitations in that they visualize only a single slice or a relatively small volume of the cell, respectively. Focused ion beam-scanning electron microscopy (FIB-SEM) has demonstrated the ability to image small volumes of cellular samples with 4-nm isotropic voxels1. Owing to advances in the precision and stability of FIB milling, together with enhanced signal detection and faster SEM scanning, we have increased the volume that can be imaged with 4-nm voxels by two orders of magnitude. Here we present a volume EM atlas at such resolution comprising ten three-dimensional datasets for whole cells and tissues, including cancer cells, immune cells, mouse pancreatic islets and Drosophila neural tissues. These open access data (via OpenOrganelle2) represent the foundation of a field of high-resolution whole-cell volume EM and subsequent analyses, and we invite researchers to explore this atlas and pose questions.


Subject(s)
Datasets as Topic , Information Dissemination , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Cell Line , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Interphase , Islets of Langerhans/cytology , Male , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Microtubules/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Open Access Publishing , Ovarian Neoplasms/immunology , Ovarian Neoplasms/ultrastructure , Ribosomes/ultrastructure , Synaptic Vesicles/ultrastructure , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
14.
Nature ; 591(7851): 659-664, 2021 03.
Article in English | MEDLINE | ID: mdl-33658713

ABSTRACT

Symmetric cell division requires the even partitioning of genetic information and cytoplasmic contents between daughter cells. Whereas the mechanisms coordinating the segregation of the genome are well known, the processes that ensure organelle segregation between daughter cells remain less well understood1. Here we identify multiple actin assemblies with distinct but complementary roles in mitochondrial organization and inheritance in mitosis. First, we find a dense meshwork of subcortical actin cables assembled throughout the mitotic cytoplasm. This network scaffolds the endoplasmic reticulum and organizes three-dimensional mitochondrial positioning to ensure the equal segregation of mitochondrial mass at cytokinesis. Second, we identify a dynamic wave of actin filaments reversibly assembling on the surface of mitochondria during mitosis. Mitochondria sampled by this wave are enveloped within actin clouds that can spontaneously break symmetry to form elongated comet tails. Mitochondrial comet tails promote randomly directed bursts of movement that shuffle mitochondrial position within the mother cell to randomize inheritance of healthy and damaged mitochondria between daughter cells. Thus, parallel mechanisms mediated by the actin cytoskeleton ensure both equal and random inheritance of mitochondria in symmetrically dividing cells.


Subject(s)
Actins/chemistry , Actins/metabolism , Mitochondria/metabolism , Mitosis , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Animals , Cell Division , Cell Line , Cytokinesis , Endoplasmic Reticulum/metabolism , Hippocampus/cytology , Hippocampus/embryology , Humans , Mitochondria/chemistry , Neurons , Rats
15.
Proc Natl Acad Sci U S A ; 121(23): e2308531121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805288

ABSTRACT

Many animals exhibit remarkable colors that are produced by the constructive interference of light reflected from arrays of intracellular guanine crystals. These animals can fine-tune their crystal-based structural colors to communicate with each other, regulate body temperature, and create camouflage. While it is known that these changes in color are caused by changes in the angle of the crystal arrays relative to incident light, the cellular machinery that drives color change is not understood. Here, using a combination of 3D focused ion beam scanning electron microscopy (FIB-SEM), micro-focused X-ray diffraction, superresolution fluorescence light microscopy, and pharmacological perturbations, we characterized the dynamics and 3D cellular reorganization of crystal arrays within zebrafish iridophores during norepinephrine (NE)-induced color change. We found that color change results from a coordinated 20° tilting of the intracellular crystals, which alters both crystal packing and the angle at which impinging light hits the crystals. Importantly, addition of the dynein inhibitor dynapyrazole-a completely blocked this NE-induced red shift by hindering crystal dynamics upon NE addition. FIB-SEM and microtubule organizing center (MTOC) mapping showed that microtubules arise from two MTOCs located near the poles of the iridophore and run parallel to, and in between, individual crystals. This suggests that dynein drives crystal angle change in response to NE by binding to the limiting membrane surrounding individual crystals and walking toward microtubule minus ends. Finally, we found that intracellular cAMP regulates the color change process. Together, our results provide mechanistic insight into the cellular machinery that drives structural color change.


Subject(s)
Zebrafish , Animals , Norepinephrine/metabolism , Norepinephrine/pharmacology , Color , Pigmentation/physiology , Microscopy, Electron, Scanning , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/chemistry
16.
Annu Rev Biochem ; 80: 327-32, 2011.
Article in English | MEDLINE | ID: mdl-21513458

ABSTRACT

Understanding how cells of all types sense external and internal signals and how these signals are processed to yield particular responses is a major goal of biology. Genetically encoded fluorescent proteins (FPs) and fluorescent sensors are playing an important role in achieving this comprehensive knowledge base of cell function. Providing high sensitivity and immense versatility while being minimally perturbing to a biological specimen, the probes can be used in different microscopy techniques to visualize cellular processes on many spatial scales. Three review articles in this volume discuss recent advances in probe design and applications. These developments help expand the range of biochemical processes in living systems suitable for study. They provide researchers with exciting new tools to explore how cellular processes are organized and their activity regulated in vivo.


Subject(s)
Cell Physiological Phenomena , Microscopy, Fluorescence/methods , Fluorescence Resonance Energy Transfer/methods , Luminescent Proteins/metabolism , Nitric Oxide/chemistry , Signal Transduction/physiology , Zinc/chemistry
17.
Trends Biochem Sci ; 46(7): 535-549, 2021 07.
Article in English | MEDLINE | ID: mdl-33579564

ABSTRACT

Liquid-liquid phase separation (LLPS) has emerged in recent years as an important physicochemical process for organizing diverse processes within cells via the formation of membraneless organelles termed biomolecular condensates. Emerging evidence now suggests that the formation and regulation of biomolecular condensates are also intricately linked to cancer formation and progression. We review the most recent literature linking the existence and/or dissolution of biomolecular condensates to different hallmarks of cancer formation and progression. We then discuss the opportunities that this condensate perspective provides for cancer research and the development of novel therapeutic approaches, including the perturbation of condensates by small-molecule inhibitors.


Subject(s)
Neoplasms , Organelles , Humans
18.
Cell ; 141(4): 656-67, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20478256

ABSTRACT

Starvation-induced autophagosomes engulf cytosol and/or organelles and deliver them to lysosomes for degradation, thereby resupplying depleted nutrients. Despite advances in understanding the molecular basis of this process, the membrane origin of autophagosomes remains unclear. Here, we demonstrate that, in starved cells, the outer membrane of mitochondria participates in autophagosome biogenesis. The early autophagosomal marker, Atg5, transiently localizes to punctae on mitochondria, followed by the late autophagosomal marker, LC3. The tail-anchor of an outer mitochondrial membrane protein also labels autophagosomes and is sufficient to deliver another outer mitochondrial membrane protein, Fis1, to autophagosomes. The fluorescent lipid NBD-PS (converted to NBD-phosphotidylethanolamine in mitochondria) transfers from mitochondria to autophagosomes. Photobleaching reveals membranes of mitochondria and autophagosomes are transiently shared. Disruption of mitochondria/ER connections by mitofusin2 depletion dramatically impairs starvation-induced autophagy. Mitochondria thus play a central role in starvation-induced autophagy, contributing membrane to autophagosomes.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Phagosomes/metabolism , Animals , Cell Line , Cell Physiological Phenomena , Culture Media , GTP Phosphohydrolases , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Rats
19.
Proc Natl Acad Sci U S A ; 119(48): e2202580119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36417438

ABSTRACT

Neurons in the developing brain undergo extensive structural refinement as nascent circuits adopt their mature form. This physical transformation of neurons is facilitated by the engulfment and degradation of axonal branches and synapses by surrounding glial cells, including microglia and astrocytes. However, the small size of phagocytic organelles and the complex, highly ramified morphology of glia have made it difficult to define the contribution of these and other glial cell types to this crucial process. Here, we used large-scale, serial section transmission electron microscopy (TEM) with computational volume segmentation to reconstruct the complete 3D morphologies of distinct glial types in the mouse visual cortex, providing unprecedented resolution of their morphology and composition. Unexpectedly, we discovered that the fine processes of oligodendrocyte precursor cells (OPCs), a population of abundant, highly dynamic glial progenitors, frequently surrounded small branches of axons. Numerous phagosomes and phagolysosomes (PLs) containing fragments of axons and vesicular structures were present inside their processes, suggesting that OPCs engage in axon pruning. Single-nucleus RNA sequencing from the developing mouse cortex revealed that OPCs express key phagocytic genes at this stage, as well as neuronal transcripts, consistent with active axon engulfment. Although microglia are thought to be responsible for the majority of synaptic pruning and structural refinement, PLs were ten times more abundant in OPCs than in microglia at this stage, and these structures were markedly less abundant in newly generated oligodendrocytes, suggesting that OPCs contribute substantially to the refinement of neuronal circuits during cortical development.


Subject(s)
Neocortex , Oligodendrocyte Precursor Cells , Animals , Mice , Axons/metabolism , Oligodendroglia/metabolism , Neurons/metabolism
20.
Immunity ; 42(5): 864-76, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25992860

ABSTRACT

Cytotoxic T lymphocytes (CTLs) use polarized secretion to rapidly destroy virally infected and tumor cells. To understand the temporal relationships between key events leading to secretion, we used high-resolution 4D imaging. CTLs approached targets with actin-rich projections at the leading edge, creating an initially actin-enriched contact with rearward-flowing actin. Within 1 min, cortical actin reduced across the synapse, T cell receptors (TCRs) clustered centrally to form the central supramolecular activation cluster (cSMAC), and centrosome polarization began. Granules clustered around the moving centrosome within 2.5 min and reached the synapse after 6 min. TCR-bearing intracellular vesicles were delivered to the cSMAC as the centrosome docked. We found that the centrosome and granules were delivered to an area of membrane with reduced cortical actin density and phospholipid PIP2. These data resolve the temporal order of events during synapse maturation in 4D and reveal a critical role for actin depletion in regulating secretion.


Subject(s)
Actins/metabolism , Cytoplasmic Granules/metabolism , Immunological Synapses/metabolism , T-Lymphocytes, Cytotoxic/cytology , Cell Membrane/chemistry , Cells, Cultured , Cytoplasmic Granules/chemistry , Fluorescent Antibody Technique , Humans , Models, Immunological , Phospholipids/metabolism , T-Lymphocytes, Cytotoxic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL