Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.243
Filter
1.
J Cell Biol ; 223(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38478018

ABSTRACT

The essential Golgi protein Sly1 is a member of the Sec1/mammalian Unc-18 (SM) family of SNARE chaperones. Sly1 was originally identified through remarkable gain-of-function alleles that bypass requirements for diverse vesicle tethering factors. Employing genetic analyses and chemically defined reconstitutions of ER-Golgi fusion, we discovered that a loop conserved among Sly1 family members is not only autoinhibitory but also acts as a positive effector. An amphipathic lipid packing sensor (ALPS)-like helix within the loop directly binds high-curvature membranes. Membrane binding is required for relief of Sly1 autoinhibition and also allows Sly1 to directly tether incoming vesicles to the Qa-SNARE on the target organelle. The SLY1-20 mutation bypasses requirements for diverse tethering factors but loses this ability if the tethering activity is impaired. We propose that long-range tethers, including Golgins and multisubunit tethering complexes, hand off vesicles to Sly1, which then tethers at close range to initiate trans-SNARE complex assembly and fusion in the early secretory pathway.


Subject(s)
Cytoplasmic Vesicles , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Animals , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Mammals/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Munc18 Proteins/analysis , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism
2.
Cell ; 187(5): 1109-1126.e21, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38382525

ABSTRACT

Oocytes are among the longest-lived cells in the body and need to preserve their cytoplasm to support proper embryonic development. Protein aggregation is a major threat for intracellular homeostasis in long-lived cells. How oocytes cope with protein aggregation during their extended life is unknown. Here, we find that mouse oocytes accumulate protein aggregates in specialized compartments that we named endolysosomal vesicular assemblies (ELVAs). Combining live-cell imaging, electron microscopy, and proteomics, we found that ELVAs are non-membrane-bound compartments composed of endolysosomes, autophagosomes, and proteasomes held together by a protein matrix formed by RUFY1. Functional assays revealed that in immature oocytes, ELVAs sequester aggregated proteins, including TDP-43, and degrade them upon oocyte maturation. Inhibiting degradative activity in ELVAs leads to the accumulation of protein aggregates in the embryo and is detrimental for embryo survival. Thus, ELVAs represent a strategy to safeguard protein homeostasis in long-lived cells.


Subject(s)
Cytoplasmic Vesicles , Oocytes , Protein Aggregates , Animals , Female , Mice , Autophagosomes , Cytoplasmic Vesicles/metabolism , Lysosomes/metabolism , Oocytes/cytology , Oocytes/metabolism , Proteasome Endopeptidase Complex , Proteolysis
3.
J Biol Chem ; 299(8): 104945, 2023 08.
Article in English | MEDLINE | ID: mdl-37348560

ABSTRACT

Human Flower (hFWE) isoforms hFWE1-4 are putative transmembrane (TM) proteins that reportedly mediate fitness comparisons during cell competition through extracellular display of their C-terminal tails. Isoform topology, subcellular localization, and duration of plasma membrane presentation are essential to this function. However, disagreement persists regarding the structure of orthologous fly and mouse FWEs, and experimental evidence for hFWE isoform subcellular localization or membrane structure is lacking. Here, we used AlphaFold2 and subsequent molecular dynamics-based structural predictions to construct epitope-tagged hFWE3 and hFWE4, the most abundant human isoforms, for experimental determination of their structure and internalization dynamics. We demonstrate that hFWE3 resides in the membrane of the endoplasmic reticulum (ER), while hFWE4 partially colocalizes with Rab4-, Rab5-, and Rab11-positive vesicles as well as with the plasma membrane. An array of imaging techniques revealed that hFWE4 positions both N- and C-terminal tails and a loop between second and third TM segments within the cytosol, while small (4-12aa) loops between the first and second and the third and fourth TM segments are either exposed to the extracellular space or within the lumen of cytoplasmic vesicles. Similarly, we found hFWE3 positions both N- and C-terminal tails in the cytosol, while a short loop between TM domains extends into the ER lumen. Finally, we demonstrate that hFWE4 exists only transiently at the cell surface and is rapidly internalized in an AP-2- and dynamin-1-dependent manner. Collectively, these data are consistent with a conserved role for hFWE4 in endocytic processes.


Subject(s)
Endoplasmic Reticulum , Models, Molecular , Humans , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endocytosis , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Molecular Dynamics Simulation , Protein Structure, Tertiary , Clathrin/metabolism , HEK293 Cells
4.
J Cell Sci ; 135(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-36052645

ABSTRACT

Primary cilia are near-ubiquitously assembled on cells in the human body, and are broadly associated with genetic diseases and cancers. In the early stage of ciliogenesis, the ciliary vesicle (CV) is formed on the mother centriole, which nucleates the primary cilium. However, the regulatory mechanisms underlying CV formation have not yet been fully elucidated. Here, we found that the atypical small GTPase RAB-like 3 (RABL3) is necessary to assemble primary cilia in human cells. RABL3 directly interacts with RAB11 (herein referring to both RAB11A and RAB11B), which is involved in CV formation. RABL3 localizes around the centrosome during early ciliogenesis, reminiscent of RAB11 dynamics. Furthermore, RABL3 positively controls the CV formation like RAB11. These findings suggest that RABL3 plays an important role, in cooperation with RAB11, in CV formation during early ciliogenesis.


Subject(s)
Cilia , rab GTP-Binding Proteins , Centrioles/metabolism , Centrosome/metabolism , Cilia/metabolism , Cytoplasmic Vesicles/metabolism , Humans , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
5.
Traffic ; 23(7): 360-373, 2022 07.
Article in English | MEDLINE | ID: mdl-35510564

ABSTRACT

The endocytic protein EHD1 controls primary ciliogenesis by facilitating fusion of the ciliary vesicle and by removal of CP110 from the mother centriole. EHD3, the closest EHD1 paralog, has a similar regulatory role, but initial evidence suggested that the other two more distal paralogs, EHD2 and EHD4 may be dispensable for ciliogenesis. Herein, we define a novel role for EHD4, but not EHD2, in regulating primary ciliogenesis. To better understand the mechanisms and differential functions of the EHD proteins in ciliogenesis, we first demonstrated a requirement for EHD1 ATP-binding to promote ciliogenesis. We then identified two sequence motifs that are entirely conserved between EH domains of EHD1, EHD3 and EHD4, but display key amino acid differences within the EHD2 EH domain. Substitution of either P446 or E470 in EHD1 with the aligning S451 or W475 residues from EHD2 was sufficient to prevent rescue of ciliogenesis in EHD1-depleted cells upon reintroduction of EHD1. Overall, our data enhance the current understanding of the EHD paralogs in ciliogenesis, demonstrate a need for ATP-binding and identify conserved sequences in the EH domains of EHD1, EHD3 and EHD4 that regulate EHD1 binding to proteins and its ability to rescue ciliogenesis in EHD1-depleted cells.


Subject(s)
Carrier Proteins , Cytoplasmic Vesicles , Adenosine Triphosphate , Animals , Carrier Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Mammals/metabolism
6.
Methods Mol Biol ; 2431: 239-247, 2022.
Article in English | MEDLINE | ID: mdl-35412280

ABSTRACT

Eukaryotic cells use microtubule-based vesicle transport to exchange molecules between compartments. Kinesin family members mediate all microtubule plus end-directed vesicle transport. Of the 45 kinesins expressed in humans, some 20 mediate microtubule plus-end directed vesicle transport. Here we describe a technique to visualize vesicle-bound kinesins in cultured hippocampal neurons. The method involves the expression of the vesicle-binding tail domain while minimizing the cytoplasmic pool. Using this approach drastically improves vesicle labeling compared to full-length kinesins. This tool is useful for systematically comparing the localization of different kinesins in the same cell type and for identifying cargo proteins that reside in vesicles moved by a specific kinesin family member. While we describe the assay in cultured hippocampal neurons, we expect it to be easily transferable to other eukaryotic cell types.


Subject(s)
Kinesins , Neurons , Cytoplasmic Vesicles/metabolism , Hippocampus/metabolism , Humans , Kinesins/metabolism , Microscopy, Fluorescence/methods , Microtubules/metabolism , Neurons/metabolism , Organelles/metabolism
7.
PLoS One ; 17(3): e0264521, 2022.
Article in English | MEDLINE | ID: mdl-35294476

ABSTRACT

Through the integration of results from an imaging analysis of intracellular trafficking of labelled neurosecretory vesicles in chromaffin cells, we develop a Markov state model to describe their transport and binding kinetics. Our simulation results indicate that a spatial redistribution of neurosecretory vesicles occurs upon secretagogue stimulation leading vesicles to the plasma membrane where they undergo fusion thereby releasing adrenaline and noradrenaline. Furthermore, we find that this redistribution alone can explain the observed up-regulation of vesicle transport upon stimulation and its directional bias towards the plasma membrane. Parameter fitting indicates that in the deeper compartment within the cell, vesicle transport is asymmetric and characterised by a bias towards the plasma membrane.


Subject(s)
Chromaffin Cells , Biological Transport , Cell Membrane/metabolism , Chromaffin Cells/metabolism , Cytoplasmic Vesicles/metabolism , Kinetics
8.
Sci Rep ; 12(1): 3003, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194033

ABSTRACT

Bacterial kidney disease (BKD) is a chronic bacterial disease affecting both wild and farmed salmonids. The causative agent for BKD is the Gram-positive fish pathogen Renibacterium salmoninarum. As treatment and prevention of BKD have proven to be difficult, it is important to know and identify the key bacterial proteins that interact with the host. We used subcellular fractionation to report semi-quantitative data for the cytosolic, membrane, extracellular, and membrane vesicle (MV) proteome of R. salmoninarum. These data can aid as a backbone for more targeted experiments regarding the development of new drugs for the treatment of BKD. Further analysis was focused on the MV proteome, where both major immunosuppressive proteins P57/Msa and P22 and proteins involved in bacterial adhesion were found in high abundance. Interestingly, the P22 protein was relatively enriched only in the extracellular and MV fraction, implicating that MVs may play a role in host-pathogen interaction. Compared to the other subcellular fractions, the MVs were also relatively enriched in lipoproteins and all four cell wall hydrolases belonging to the New Lipoprotein C/Protein of 60 kDa (NlpC/P60) family were detected, suggesting an involvement in the formation of the MVs.


Subject(s)
Cytoplasmic Vesicles/physiology , Proteome/genetics , Proteomics , Virulence , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Cytoplasmic Vesicles/metabolism , Fish Diseases/microbiology , Fishes/microbiology , Host-Parasite Interactions , Kidney Diseases/microbiology , Kidney Diseases/veterinary , Lipoproteins/metabolism , Renibacterium/cytology , Renibacterium/genetics , Renibacterium/pathogenicity , Subcellular Fractions/physiology , Virulence/genetics
9.
J Nutr Biochem ; 101: 108912, 2022 03.
Article in English | MEDLINE | ID: mdl-34801692

ABSTRACT

Glucose homeostasis imbalance and insulin resistance (IR) are major contributors to the incidence of type 2 diabetes. Omega-3 polyunsaturated fatty acids (PUFAs) are key ingredients for maintaining cellular functions and improving insulin sensitivity. However, how omega-3 PUFAs modulate the dynamic process of glucose transport at the cellular level remains unclear. Here we unraveled eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may regulate the glucose transporter 4 (GLUT4) vesicle trafficking in both normal and IR adipocytes. Both omega-3 PUFAs significantly increase glucose consumption within a range of 10-32% in the basal state. Furthermore, both EPA (200 µM) and DHA (100 µM) may significantly promote the serine/threonine protein kinase (Akt) phosphorylation by 70% and 40% in the physiological state of adipocytes, respectively. Both omega-3 PUFAs significantly advanced the Akt phosphorylation in a dose-dependent way and showed a ∼2-fold increase at the dose of 200 µM in the IR pathological state. However, they could not up-regulate the expression of GLUT4 and insulin-regulated aminopeptidase protein. We further revealed that both omega-3 PUFAs dynamically promote insulin-stimulated GLUT4 vesicle translocation and soluble N-ethylmaleimide-sensitive factor attachment protein receptor mediated vesicle docking and fusion to the plasma membrane via specifically modulating the expression of vesicle-associated membrane protein 2. Understanding the mechanisms by which omega-3 PUFAs modulate cellular metabolism and IR in peripheral tissues may provide novel insights into the potential impact of omega-3 PUFAs on the metabolic function and the management of IR.


Subject(s)
Adipocytes/metabolism , Fatty Acids, Omega-3/pharmacology , Glucose Transporter Type 4/metabolism , SNARE Proteins/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Glucose/metabolism , Insulin/metabolism , Insulin Resistance , Mice , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
10.
Int J Mol Sci ; 22(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830133

ABSTRACT

The primary cilium is found in most mammalian cells and plays a functional role in tissue homeostasis and organ development by modulating key signaling pathways. Ciliopathies are a group of genetically heterogeneous disorders resulting from defects in cilia development and function. Patients with ciliopathic disorders exhibit a range of phenotypes that include nephronophthisis (NPHP), a progressive tubulointerstitial kidney disease that commonly results in end-stage renal disease (ESRD). In recent years, distal appendages (DAPs), which radially project from the distal end of the mother centriole, have been shown to play a vital role in primary ciliary vesicle docking and the initiation of ciliogenesis. Mutations in the genes encoding these proteins can result in either a complete loss of the primary cilium, abnormal ciliary formation, or defective ciliary signaling. DAPs deficiency in humans or mice commonly results in NPHP. In this review, we outline recent advances in our understanding of the molecular functions of DAPs and how they participate in nephronophthisis development.


Subject(s)
Centrosome/metabolism , Cilia/metabolism , Kidney Diseases, Cystic/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Basal Bodies/metabolism , Cell Membrane/metabolism , Centrioles/metabolism , Cytoplasmic Vesicles/metabolism , Humans , Kidney Diseases, Cystic/congenital , Models, Biological
12.
J Mol Biol ; 433(24): 167323, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34695381

ABSTRACT

Misfolding and aggregation of alpha-synuclein (αS) within dopaminergic neurons is a key factor in the development and progression of a group of age-related neurodegenerative diseases, termed synucleinopathies, that include Parkinson's disease (PD). We previously derived a peptide inhibitor from a 209,952-member intracellular library screen by employing the preNAC region (45-54) as a design template. At least six single-point mutations firmly linked to early-onset Parkinson's disease (E46K, H50Q, G51D, A53T/E/V) are located within this region, strongly implicating a pathogenic role within αS that leads to increased cytotoxicity. A library-derived ten residue peptide, 4554W, was consequently shown to block αS aggregation at the point of primary nucleation via lipid induction, inhibiting its conversion into downstream cytotoxic species. Here we couple truncation with a full alanine scan analysis, to establish the effect upon the αS aggregation pathway relative to 4554W. This revealed the precise residues responsible for eliciting inhibitory interaction and function, as well as those potentially amenable to modification or functionalisation. We find that modification N6A combined with N-terminal truncation results in a peptide of significantly increased efficacy. Importantly, our data demonstrate that the peptide does not directly disrupt αS lipid-binding, a desirable trait since antagonists of αS aggregation and toxicity should not impede association with small synaptic neurotransmitter vesicles, and thus not disrupt dopaminergic vesicle fusion and recycling. This work paves the way toward the major aim of deriving a highly potent peptide antagonist of αS pathogenicity without impacting on native αS function.


Subject(s)
Antiparkinson Agents/chemistry , Parkinson Disease/metabolism , Peptidomimetics/chemistry , Protein Aggregates/drug effects , Protein Folding/drug effects , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry , Alanine/chemistry , Alanine/genetics , Antiparkinson Agents/pharmacology , Cryoelectron Microscopy , Cytoplasmic Vesicles/metabolism , Dopaminergic Neurons/metabolism , Humans , Lipids/chemistry , Parkinson Disease/genetics , Peptide Library , Peptidomimetics/pharmacology , Point Mutation , alpha-Synuclein/genetics
13.
J Cell Biol ; 220(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34714326

ABSTRACT

Mechanisms that turn over components of the nucleus and inner nuclear membrane (INM) remain to be fully defined. We explore how components of the INM are selected by a cytosolic autophagy apparatus through a transmembrane nuclear envelope-localized cargo adaptor, Atg39. A split-GFP reporter showed that Atg39 localizes to the outer nuclear membrane (ONM) and thus targets the INM across the nuclear envelope lumen. Consistent with this, sequence elements that confer both nuclear envelope localization and a membrane remodeling activity are mapped to the Atg39 lumenal domain; these lumenal motifs are required for the autophagy-mediated degradation of integral INM proteins. Interestingly, correlative light and electron microscopy shows that the overexpression of Atg39 leads to the expansion of the ONM and the enclosure of a network of INM-derived vesicles in the nuclear envelope lumen. Thus, we propose an outside-in model of nucleophagy where INM is delivered into vesicles in the nuclear envelope lumen, which can be targeted by the autophagosome.


Subject(s)
Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Nuclear Envelope/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Autophagosomes/ultrastructure , Autophagy , Autophagy-Related Proteins/chemistry , Cytoplasmic Vesicles/ultrastructure , Green Fluorescent Proteins/metabolism , Nuclear Envelope/ultrastructure , Protein Domains , Receptors, Cytoplasmic and Nuclear/chemistry , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/chemistry , Structure-Activity Relationship , Time Factors , Vacuoles/metabolism , Vacuoles/ultrastructure , Vesicular Transport Proteins/metabolism
14.
Biomolecules ; 11(9)2021 09 09.
Article in English | MEDLINE | ID: mdl-34572546

ABSTRACT

Abnormal accumulation of the protein α- synuclein (α-syn) into proteinaceous inclusions called Lewy bodies (LB) is the neuropathological hallmark of Parkinson's disease (PD) and related disorders. Interestingly, a growing body of evidence suggests that LB are also composed of other cellular components such as cellular membrane fragments and vesicular structures, suggesting that dysfunction of the endolysosomal system might also play a role in LB formation and neuronal degeneration. Yet the link between α-syn aggregation and the endolysosomal system disruption is not fully elucidated. In this review, we discuss the potential interaction between α-syn and the endolysosomal system and its impact on PD pathogenesis. We propose that the accumulation of monomeric and aggregated α-syn disrupt vesicles trafficking, docking, and recycling, leading to the impairment of the endolysosomal system, notably the autophagy-lysosomal degradation pathway. Reciprocally, PD-linked mutations in key endosomal/lysosomal machinery genes (LRRK2, GBA, ATP13A2) also contribute to increasing α-syn aggregation and LB formation. Altogether, these observations suggest a potential synergistic role of α-syn and the endolysosomal system in PD pathogenesis and represent a viable target for the development of disease-modifying treatment for PD and related disorders.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Cytoplasmic Vesicles/metabolism , Humans , Proteolysis
15.
Front Immunol ; 12: 662803, 2021.
Article in English | MEDLINE | ID: mdl-34381444

ABSTRACT

Phagocytosis is the cellular defense mechanism used to eliminate antigens derived from dysregulated or damaged cells, and microbial pathogens. Phagocytosis is therefore a pillar of innate immunity, whereby foreign particles are engulfed and degraded in lysolitic vesicles. In hexacorallians, phagocytic mechanisms are poorly understood, though putative anthozoan phagocytic cells (amoebocytes) have been identified histologically. We identify and characterize phagocytes from the coral Pocillopora damicornis and the sea anemone Nematostella vectensis. Using fluorescence-activated cell sorting and microscopy, we show that distinct populations of phagocytic cells engulf bacteria, fungal antigens, and beads. In addition to pathogenic antigens, we show that phagocytic cells engulf self, damaged cells. We show that target antigens localize to low pH phagolysosomes, and that degradation is occurring within them. Inhibiting actin filament rearrangement interferes with efficient particle phagocytosis but does not affect small molecule pinocytosis. We also demonstrate that cellular markers for lysolitic vesicles and reactive oxygen species (ROS) correlate with hexacorallian phagocytes. These results establish a foundation for improving our understanding of hexacorallian immune cell biology.


Subject(s)
Anthozoa/immunology , Phagocytes/immunology , Animals , Anthozoa/metabolism , Biomarkers , Cytokines/metabolism , Cytoplasmic Vesicles/metabolism , Flow Cytometry , Hydrogen-Ion Concentration , Immunity, Innate , Phagocytes/cytology , Phagocytes/metabolism , Phagocytosis/immunology , Phagosomes , Sea Anemones
16.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34260378

ABSTRACT

Centrosome duplication and DNA replication are two pivotal events that higher eukaryotic cells use to initiate proliferation. While DNA replication is initiated through origin licensing, centrosome duplication starts with cartwheel assembly and is partly controlled by CP110. However, the upstream coordinator for both events has been, until now, a mystery. Here, we report that suppressor of fused protein (Sufu), a negative regulator of the Hedgehog (Hh) pathway playing a significant role in restricting the trafficking and function of glioma-related (Gli) proteins, acts as an upstream switch by facilitating CP110 phosphorylation by CDK2, promoting intranuclear Cdt1 degradation and excluding prereplication complex (pre-RC) components from chromosomes, independent of its canonical function in the Hh pathway. We found that Sufu localizes to both the centrosome and the nucleus and that knockout of Sufu induces abnormalities including centrosome amplification, increased nuclear size, multipolar spindle formation, and polyploidy. Serum stimulation promotes the elimination of Sufu from the centrosome by vesicle release at the ciliary tip and from the nucleus via protein degradation, which allows centrosome duplication and DNA replication to proceed. Collectively, this work reveals a mechanism through which Sufu negatively regulates the G1-S transition.


Subject(s)
Centrosome/metabolism , DNA Replication , Repressor Proteins/metabolism , Animals , Calmodulin-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Death , Cell Nucleus/metabolism , Cilia/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cytoplasmic Vesicles/metabolism , Fibroblasts/metabolism , G1 Phase , HEK293 Cells , HeLa Cells , Hedgehog Proteins/metabolism , Humans , Mice , Mitosis , Mutation/genetics , Phosphorylation , Proteolysis , Repressor Proteins/genetics , S Phase
17.
Emerg Microbes Infect ; 10(1): 1378-1389, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34162308

ABSTRACT

The vector-borne flaviviruses (VBFVs) are well known for causing great misery and death in humans worldwide. The VBFVs include those transmitted by mosquitos, such as Zika virus (ZIKV), dengue virus; and those transmitted by ticks including the tick-borne flavivirus serocomplex and Powassan virus (POWV). Two of our recent reports showed that intracranial POWV infection in the reservoir host, Peromyscus leucopus, was restricted and caused no overt clinical disease. Several modes of analyses suggested activation of the LXR pathway. Activation of the LXR pathway leads to increased efflux of cholesterol from cells and consequent disturbances in membrane biogenesis. Because VBFV replication is dependent on membrane biogenesis, we evaluated the effect of an LXR agonist (LXR623) on POWV and ZIKV infection and observed that the compound impaired permissive replication of both viruses in a human neuroblastoma SK-N-SH cell line. The LXR agonist resulted in failure of the viruses to induce ER expansion and elaborate vesicle formation, suggesting that the efflux of cholesterol was part of the antiviral mechanism. We also observed that the LXR agonist contributed to the mechanism of virus suppression by increased expression of mRNAs encoding for the antiviral cytokines CXCL10, RANTES and IFN1ß. In sharp contrast, a LXR antagonist (GSK2033) had no significant effect on VBFV replication. We conclude that LXR623 impairs flavivirus replication by stimulating cellular antiviral factors.


Subject(s)
Encephalitis Viruses, Tick-Borne/drug effects , Indazoles/pharmacology , Liver X Receptors/agonists , Zika Virus/drug effects , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/metabolism , Cytopathogenic Effect, Viral/drug effects , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Encephalitis Viruses, Tick-Borne/physiology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Liver X Receptors/metabolism , Virus Replication/drug effects , Zika Virus/physiology
18.
Mol Biol Cell ; 32(17): 1594-1610, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34161137

ABSTRACT

The Golgi complex is a central hub for intracellular protein trafficking and glycosylation. Steady-state localization of glycosylation enzymes is achieved by a combination of mechanisms involving retention and recycling, but the machinery governing these mechanisms is poorly understood. Herein we show that the Golgi-associated retrograde protein (GARP) complex is a critical component of this machinery. Using multiple human cell lines, we show that depletion of GARP subunits impairs Golgi modification of N- and O-glycans and reduces the stability of glycoproteins and Golgi enzymes. Moreover, GARP-knockout (KO) cells exhibit reduced retention of glycosylation enzymes in the Golgi. A RUSH assay shows that, in GARP-KO cells, the enzyme beta-1,4-galactosyltransferase 1 is not retained at the Golgi complex but instead is missorted to the endolysosomal system. We propose that the endosomal system is part of the trafficking itinerary of Golgi enzymes or their recycling adaptors and that the GARP complex is essential for recycling and stabilization of the Golgi glycosylation machinery. [Media: see text].


Subject(s)
Golgi Apparatus/metabolism , Protein Transport/physiology , Vesicular Transport Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Endosomes/metabolism , Glycosylation , HeLa Cells , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , trans-Golgi Network/metabolism
19.
Commun Biol ; 4(1): 678, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083706

ABSTRACT

Toxicity mechanisms of metal oxide nanoparticles towards bacteria and underlying roles of membrane composition are still debated. Herein, the response of lipopolysaccharide-truncated Escherichia coli K12 mutants to TiO2 nanoparticles (TiO2NPs, exposure in dark) is addressed at the molecular, single cell, and population levels by transcriptomics, fluorescence assays, cell nanomechanics and electrohydrodynamics. We show that outer core-free lipopolysaccharides featuring intact inner core increase cell sensitivity to TiO2NPs. TiO2NPs operate as membrane strippers, which induce osmotic stress, inactivate cell osmoregulation and initiate lipid peroxidation, which ultimately leads to genesis of membrane vesicles. In itself, truncation of lipopolysaccharide inner core triggers membrane permeabilization/depolarization, lipid peroxidation and hypervesiculation. In turn, it favors the regulation of TiO2NP-mediated changes in cell Turgor stress and leads to efficient vesicle-facilitated release of damaged membrane components. Remarkably, vesicles further act as electrostatic baits for TiO2NPs, thereby mitigating TiO2NPs toxicity. Altogether, we highlight antagonistic lipopolysaccharide-dependent bacterial responses to nanoparticles and we show that the destabilized membrane can generate unexpected resistance phenotype.


Subject(s)
Cytoplasmic Vesicles/drug effects , Escherichia coli/drug effects , Metal Nanoparticles/toxicity , Osmotic Pressure/drug effects , Titanium/toxicity , Cytoplasmic Vesicles/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Bacterial/drug effects , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Microscopy, Atomic Force/methods , Mutation
20.
ACS Synth Biol ; 10(6): 1284-1291, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34081866

ABSTRACT

Minicells are nanosized membrane vesicles produced by bacteria. Minicells are chromosome-free but contain cellular biosynthetic and metabolic machinery, and they are robust due to the protection provided by the bacterial cell envelope, which makes them potentially highly attractive in biomedical applications. However, the applicability of minicells and other nanoparticle-based delivery systems is limited by their inefficient accumulation at the target. Here we engineered the minicell-producing Escherichia coli strain to overexpress flagellar genes, which enables the generation of motile minicells. We subsequently performed an experimental and theoretical analysis of the minicell motility and their responses to gradients of chemoeffectors. Despite important differences between the motility of minicells and normal bacterial cells, minicells were able to bias their movement in chemical gradients and to accumulate toward the sources of chemoattractants. Such motile and chemotactic minicells may thus be applicable for an active effector delivery and specific targeting of tissues and cells according to their metabolic profiles.


Subject(s)
Chemotaxis/genetics , Cytoplasmic Vesicles/metabolism , Drug Delivery Systems/methods , Escherichia coli/metabolism , Nanoparticles/metabolism , Chemotactic Factors , Escherichia coli/genetics , Flagella/genetics , Gene Expression , Genetic Engineering/methods , Microorganisms, Genetically-Modified , Movement , Point Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...