Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Traffic ; 24(8): 334-354, 2023 08.
Article in English | MEDLINE | ID: mdl-37218497

ABSTRACT

Previously, we found that age-dependent accumulation of beta-amyloid is not sufficient to cause synaptic decline. Late-endocytic organelles (LEOs) may be driving synaptic decline as lysosomes (Lys) are a target of cellular aging and relevant for synapses. We found that LAMP1-positive LEOs increased in size and number and accumulated near synapses in aged neurons and brains. LEOs' distal accumulation might relate to the increased anterograde movement in aged neurons. Dissecting the LEOs, we found that late-endosomes accumulated while there are fewer terminal Lys in aged neurites, but not in the cell body. The most abundant LEOs were degradative Lys or endolysosomes (ELys), especially in neurites. ELys activity was reduced because of acidification defects, supported by the reduction in v-ATPase subunit V0a1 with aging. Increasing the acidification of aged ELys recovered degradation and reverted synaptic decline, while alkalinization or v-ATPase inhibition, mimicked age-dependent Lys and synapse dysfunction. We identify ELys deacidification as a neuronal mechanism of age-dependent synapse loss. Our findings suggest that future therapeutic strategies to address endolysosomal defects might be able to delay age-related synaptic decline.


Subject(s)
Neurons , Synapses , Neurons/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Adenosine Triphosphatases/metabolism
2.
Nat Commun ; 14(1): 1918, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024503

ABSTRACT

Parkinson's Disease (PD) is a common neurodegenerative disorder affecting millions of people worldwide for which there are only symptomatic therapies. Small molecules able to target key pathological processes in PD have emerged as interesting options for modifying disease progression. We have previously shown that a (poly)phenol-enriched fraction (PEF) of Corema album L. leaf extract modulates central events in PD pathogenesis, namely α-synuclein (αSyn) toxicity, aggregation and clearance. PEF was now subjected to a bio-guided fractionation with the aim of identifying the critical bioactive compound. We identified genipin, an iridoid, which relieves αSyn toxicity and aggregation. Furthermore, genipin promotes metabolic alterations and modulates lipid storage and endocytosis. Importantly, genipin was able to prevent the motor deficits caused by the overexpression of αSyn in a Drosophila melanogaster model of PD. These findings widens the possibility for the exploitation of genipin for PD therapeutics.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , Drosophila melanogaster/metabolism , Parkinson Disease/metabolism , Iridoids/pharmacology , Phenols , Lipids
3.
Cells ; 11(19)2022 09 27.
Article in English | MEDLINE | ID: mdl-36230978

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant disease caused by a CTG repeat expansion in the 3' untranslated region of the dystrophia myotonica protein kinase gene. AKT dephosphorylation and autophagy are associated with DM1. Autophagy has been widely studied in DM1, although the endocytic pathway has not. AKT has a critical role in endocytosis, and its phosphorylation is mediated by the activation of tyrosine kinase receptors, such as epidermal growth factor receptor (EGFR). EGF-activated EGFR triggers the internalization and degradation of ligand-receptor complexes that serve as a PI3K/AKT signaling platform. Here, we used primary fibroblasts from healthy subjects and DM1 patients. DM1-derived fibroblasts showed increased autophagy flux, with enlarged endosomes and lysosomes. Thereafter, cells were stimulated with a high concentration of EGF to promote EGFR internalization and degradation. Interestingly, EGF binding to EGFR was reduced in DM1 cells and EGFR internalization was also slowed during the early steps of endocytosis. However, EGF-activated EGFR enhanced AKT and ERK1/2 phosphorylation levels in the DM1-derived fibroblasts. Therefore, there was a delay in EGF-stimulated EGFR endocytosis in DM1 cells; this alteration might be due to the decrease in the binding of EGF to EGFR, and not to a decrease in AKT phosphorylation.


Subject(s)
Epidermal Growth Factor , Myotonic Dystrophy , 3' Untranslated Regions , Epidermal Growth Factor/genetics , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Ligands , Myotonic Dystrophy/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism
4.
BMC Biol ; 20(1): 111, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35549945

ABSTRACT

BACKGROUND: In vertebrate cells, the Golgi functional subunits, mini-stacks, are linked into a tri-dimensional network. How this "ribbon" architecture relates to Golgi functions remains unclear. Are all connections between mini-stacks equal? Is the local structure of the ribbon of functional importance? These are difficult questions to address, without a quantifiable readout of the output of ribbon-embedded mini-stacks. Endothelial cells produce secretory granules, the Weibel-Palade bodies (WPB), whose von Willebrand Factor (VWF) cargo is central to hemostasis. The Golgi apparatus controls WPB size at both mini-stack and ribbon levels. Mini-stack dimensions delimit the size of VWF "boluses" whilst the ribbon architecture allows their linear co-packaging, thereby generating WPBs of different lengths. This Golgi/WPB size relationship suits mathematical analysis. RESULTS: WPB lengths were quantized as multiples of the bolus size and mathematical modeling simulated the effects of different Golgi ribbon organizations on WPB size, to be compared with the ground truth of experimental data. An initial simple model, with the Golgi as a single long ribbon composed of linearly interlinked mini-stacks, was refined to a collection of mini-ribbons and then to a mixture of mini-stack dimers plus long ribbon segments. Complementing these models with cell culture experiments led to novel findings. Firstly, one-bolus sized WPBs are secreted faster than larger secretory granules. Secondly, microtubule depolymerization unlinks the Golgi into equal proportions of mini-stack monomers and dimers. Kinetics of binding/unbinding of mini-stack monomers underpinning the presence of stable dimers was then simulated. Assuming that stable mini-stack dimers and monomers persist within the ribbon resulted in a final model that predicts a "breathing" arrangement of the Golgi, where monomer and dimer mini-stacks within longer structures undergo continuous linking/unlinking, consistent with experimentally observed WPB size distributions. CONCLUSIONS: Hypothetical Golgi organizations were validated against a quantifiable secretory output. The best-fitting Golgi model, accounting for stable mini-stack dimers, is consistent with a highly dynamic ribbon structure, capable of rapid rearrangement. Our modeling exercise therefore predicts that at the fine-grained level the Golgi ribbon is more complex than generally thought. Future experiments will confirm whether such a ribbon organization is endothelial-specific or a general feature of vertebrate cells.


Subject(s)
Endothelial Cells , von Willebrand Factor , Cells, Cultured , Exocytosis , Golgi Apparatus , Weibel-Palade Bodies/physiology , von Willebrand Factor/pharmacology , von Willebrand Factor/physiology
5.
Front Endocrinol (Lausanne) ; 13: 1008418, 2022.
Article in English | MEDLINE | ID: mdl-36589826

ABSTRACT

Introduction: Diabetes is one of the major metabolic diseases worldwide. Despite being a complex systemic pathology, the aggregation and deposition of Islet Amyloid Polypeptide (IAPP), or amylin, is a recognized histopathological marker of the disease. Although IAPP proteotoxicity represents an important trigger of ß-cell dysfunction and ultimately death, its exploitation as a therapeutic tool remains underdeveloped. The bioactivity of (poly)phenols towards inhibition of pathological protein aggregation is well known, however, most of the identified molecules have limited bioavailability. Methods: Using a strategy combining in silico, cell-free and cell studies, we scrutinized a unique in-house collection of (poly)phenol metabolites predicted to appear in the human circulation after (poly)phenols ingestion. Results: We identified urolithin B as a potent inhibitor of IAPP aggregation and a powerful modulator of cell homeostasis pathways. Urolithin B was shown to affect IAPP aggregation pattern, delaying the formation of amyloid fibrils and altering their size and morphology. The molecular mechanisms underlying urolithin B-mediated protection include protein clearance pathways, mitochondrial function, and cell cycle ultimately rescuing IAPP-mediated cell dysfunction and death. Discussion: In brief, our study uncovered urolithin B as a novel small molecule targeting IAPP pathological aggregation with potential to be exploited as a therapeutic tool for mitigating cellular dysfunction in diabetes. Resulting from the colonic metabolism of dietary ellagic acid in the human body, urolithin B bioactivity has the potential to be explored in nutritional, nutraceutical, and pharmacological perspectives.


Subject(s)
Diabetes Mellitus , Islet Amyloid Polypeptide , Humans , Coumarins/pharmacology , Phenols
6.
Invest Ophthalmol Vis Sci ; 62(9): 39, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34313720

ABSTRACT

Purpose: We aim to characterize the pathways required for autofluorescent granule (AFG) formation by RPE cells using cultured monolayers. Methods: We fed RPE monolayers in culture with a single pulse of photoreceptor outer segments (POS). After 24 hours the cells started accumulating AFGs that were comparable to lipofuscin in vivo. Using this model, we used a variety of light and electron microscopical techniques, flow cytometry and Western blot to analyze the formation of AFGs. We also generated a mutant RPE line lacking cathepsin D by gene editing. Results: AFGs seem to derive from incompletely digested POS-containing phagosomes and after 3 days are surrounded by a single membrane positive for lysosome markers. We show by various methods that lysosome-phagosome fusion is required for AFG formation, and that impairment of lysosomal pH or catalytic activity, particularly cathepsin D activity, enhances AF accumulation. Conclusions: We conclude that lysosomal dysfunction results in incomplete POS degradation and enhanced AFG accumulation.


Subject(s)
Lipofuscin/metabolism , Lysosomes/metabolism , Retinal Pigment Epithelium/metabolism , Rod Cell Outer Segment/metabolism , Animals , Blotting, Western , Cells, Cultured , Flow Cytometry , Humans , Models, Animal , Phagocytosis/physiology , Retinal Pigment Epithelium/cytology , Swine
7.
Dev Cell ; 49(5): 786-801.e6, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31056345

ABSTRACT

How can anterograde membrane trafficking be modulated by physiological cues? A screen of Golgi-associated proteins revealed that the ARF-GEF GBF1 can selectively modulate the ER-Golgi trafficking of prohaemostatic von Willebrand factor (VWF) and extracellular matrix (ECM) proteins in human endothelial cells and in mouse fibroblasts. The relationship between levels of GBF1 and the trafficking of VWF into forming secretory granules confirmed GBF1 is a limiting factor in this process. Further, GBF1 activation by AMPK couples its control of anterograde trafficking to physiological cues; levels of glucose control GBF1 activation in turn modulating VWF trafficking into secretory granules. GBF1 modulates both ER and TGN exit, the latter dramatically affecting the size of the VWF storage organelles, thereby influencing the hemostatic capacity of the endothelium. The role of AMPK as a central integrating element of cellular pathways with intra- and extra-cellular cues can now be extended to modulation of the anterograde secretory pathway.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/metabolism , von Willebrand Factor/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factors/genetics , AMP-Activated Protein Kinases/genetics , Animals , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Guanine Nucleotide Exchange Factors/genetics , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Membranes/metabolism , Mice , Phosphorylation , Protein Transport , von Willebrand Factor/genetics
8.
Traffic ; 20(4): 301-304, 2019 04.
Article in English | MEDLINE | ID: mdl-30801937

ABSTRACT

The mechanisms that regulate skin pigmentation have been the subject of intense research in recent decades. In contrast with melanin biogenesis and transport within melanocytes, little is known about how melanin is transferred and processed within keratinocytes. Several models have been proposed for how melanin is transferred, with strong evidence supporting coupled exo/endocytosis. Recently, two reports suggest that upon internalization, melanin is stored within keratinocytes in an arrested compartment, allowing the pigment to persist for long periods. In this commentary, we identify a striking parallelism between melanin processing within keratinocytes and the host-pathogen interaction with Plasmodium, opening new avenues to understand the complex molecular mechanisms that ensure skin pigmentation and photoprotection.


Subject(s)
Keratinocytes , Melanins , Host-Pathogen Interactions , Melanocytes , Skin Pigmentation
9.
J Cell Sci ; 130(21): 3611-3617, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29093059

ABSTRACT

The vascular environment can rapidly alter, and the speed with which responses to both physiological and pathological changes are required necessitates the existence of a highly responsive system. The endothelium can quickly deliver bioactive molecules by regulated exocytosis of its secretory granules, the Weibel-Palade bodies (WPBs). WPBs include proteins that initiate both haemostasis and inflammation, as well those that modulate blood pressure and angiogenesis. WPB formation is driven by von Willebrand factor, their most abundant protein, which controls both shape and size of WPBs. WPB are generated in a range of sizes, with the largest granules over ten times the size of the smallest. In this Cell Science at a Glance and the accompanying poster, we discuss the emerging mechanisms by which WPB size is controlled and how this affects the ability of this organelle to modulate haemostasis. We will also outline the different modes of exocytosis and their polarity that are currently being explored, and illustrate that these large secretory organelles provide a model for how elements of secretory granule biogenesis and exocytosis cooperate to support a complex and diverse set of functions.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , Exocytosis/physiology , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Blood Vessels/cytology , Endothelial Cells/ultrastructure , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Homeostasis/physiology , Humans , Microtubules/metabolism , Microtubules/ultrastructure , Organelle Shape , Organelle Size , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction , Weibel-Palade Bodies/ultrastructure , von Willebrand Factor/genetics
10.
J Cell Sci ; 129(10): 2096-105, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27068535

ABSTRACT

Weibel-Palade bodies (WPBs) are endothelial storage organelles that mediate the release of molecules involved in thrombosis, inflammation and angiogenesis, including the pro-thrombotic glycoprotein von Willebrand factor (VWF). Although many protein components required for WPB formation and function have been identified, the role of lipids is almost unknown. We examined two key phosphatidylinositol kinases that control phosphatidylinositol 4-phosphate levels at the trans-Golgi network, the site of WPB biogenesis. RNA interference of the type II phosphatidylinositol 4-kinases PI4KIIα and PI4KIIß in primary human endothelial cells leads to formation of an increased proportion of short WPB with perturbed packing of VWF, as exemplified by increased exposure of antibody-binding sites. When stimulated with histamine, these cells release normal levels of VWF yet, under flow, form very few platelet-catching VWF strings. In PI4KIIα-deficient mice, immuno-microscopy revealed that VWF packaging is also perturbed and these mice exhibit increased blood loss after tail cut compared to controls. This is the first demonstration that lipid kinases can control the biosynthesis of VWF and the formation of WPBs that are capable of full haemostatic function.


Subject(s)
Endothelial Cells/metabolism , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Weibel-Palade Bodies/metabolism , von Willebrand Factor/genetics , Animals , Endothelial Cells/pathology , Exocytosis , Gene Expression Regulation , Histamine/administration & dosage , Humans , Inflammation/genetics , Inflammation/pathology , Lipids/genetics , Mice , Neovascularization, Pathologic/genetics , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , RNA Interference , Thrombosis/genetics , Thrombosis/pathology , Weibel-Palade Bodies/genetics , trans-Golgi Network/genetics , trans-Golgi Network/metabolism , von Willebrand Factor/biosynthesis
11.
Blood ; 128(2): 277-85, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27106123

ABSTRACT

The von Willebrand factor (VWF) synthesized and secreted by endothelial cells is central to hemostasis and thrombosis, providing a multifunctional adhesive platform that brings together components needed for these processes. VWF secretion can occur from both apical and basolateral sides of endothelial cells, and from constitutive, basal, and regulated secretory pathways, the latter two via Weibel-Palade bodies (WPB). Although the amount and structure of VWF is crucial to its function, the extent of VWF release, multimerization, and polarity of the 3 secretory pathways have only been addressed separately, and with conflicting results. We set out to clarify these relationships using polarized human umbilical vein endothelial cells (HUVECs) grown on Transwell membranes. We found that regulated secretion of ultra-large (UL)-molecular-weight VWF predominantly occurred apically, consistent with a role in localized platelet capture in the vessel lumen. We found that constitutive secretion of low-molecular-weight (LMW) VWF is targeted basolaterally, toward the subendothelial matrix, using the adaptor protein complex 1 (AP-1), where it may provide the bulk of collagen-bound subendothelial VWF. We also found that basally-secreted VWF is composed of UL-VWF, released continuously from WPBs in the absence of stimuli, and occurs predominantly apically, suggesting this could be the main source of circulating plasma VWF. Together, we provide a unified dataset reporting the amount and multimeric state of VWF secreted from the constitutive, basal, and regulated pathways in polarized HUVECs, and have established a new role for AP-1 in the basolateral constitutive secretion of VWF.


Subject(s)
Adaptor Protein Complex 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Protein Multimerization/physiology , von Willebrand Factor/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans
12.
Cell Microbiol ; 18(3): 437-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26399761

ABSTRACT

Autophagy plays an important role in the defence against intracellular pathogens. However, some microorganisms can manipulate this host cell pathway to their advantage. In this study, we addressed the role of host cell autophagy during Plasmodium berghei liver infection. We show that vesicles containing the autophagic marker LC3 surround parasites from early time-points after invasion and throughout infection and colocalize with the parasitophorous vacuole membrane. Moreover, we show that the LC3-positive vesicles that surround Plasmodium parasites are amphisomes that converge from the endocytic and autophagic pathways, because they contain markers of both pathways. When the host autophagic pathway was inhibited by silencing several of its key regulators such as LC3, Beclin1, Vps34 or Atg5, we observed a reduction in parasite size. We also found that LC3 surrounds parasites in vivo and that parasite load is diminished in a mouse model deficient for autophagy. Together, these results show the importance of the host autophagic pathway for parasite development during the liver stage of Plasmodium infection.


Subject(s)
Autophagy/physiology , Host-Parasite Interactions/physiology , Liver/parasitology , Malaria/pathology , Plasmodium berghei/pathogenicity , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Liver/pathology , Malaria/parasitology , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism
13.
PLoS One ; 8(2): e57769, 2013.
Article in English | MEDLINE | ID: mdl-23460904

ABSTRACT

The retinal pigment epithelium (RPE) is a pigmented monolayer of cells lying between the photoreceptors and a layer of fenestrated capillaries, the choriocapillaris. Choroideremia (CHM) is an X-linked progressive degeneration of these three layers caused by the loss of function of Rab Escort protein-1 (REP1). REP1 is involved in the prenylation of Rab proteins, key regulators of membrane trafficking. To study the pathological consequences of chronic disruption of membrane traffic in the RPE we used a cell type-specific knock-out mouse model of the disease, where the Chm/Rep1 gene is deleted only in pigmented cells (Chm(Flox), Tyr-Cre+). Transmission electron microscopy (TEM) was used to quantitate the melanosome distribution in the RPE and immunofluorescent staining of rhodopsin was used to quantitate phagocytosed rod outer segments in retinal sections. The ultrastructure of the RPE and Bruch's membrane at different ages was characterised by TEM to analyse age-related changes occurring as a result of defects in membrane traffic pathways. Chm/Rep1 gene knockout in RPE cells resulted in reduced numbers of melanosomes in the apical processes and delayed phagosome degradation. In addition, the RPE accumulated pathological changes at 5-6 months of age similar to those observed in 2-year old controls. These included the intracellular accumulation of lipofuscin-containing deposits, disorganised basal infoldings and the extracellular accumulation of basal laminar and basal linear deposits. The phenotype of the Chm(Flox), Tyr-Cre+ mice suggests that loss of the Chm/Rep1 gene causes premature accumulation of features of aging in the RPE. Furthermore, the striking similarities between the present observations and some of the phenotypes reported in age-related macular degeneration (AMD) suggest that membrane traffic defects may contribute to the pathogenesis of AMD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Aging/pathology , Gene Deletion , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Adaptor Proteins, Signal Transducing/metabolism , Aging/metabolism , Animals , Bruch Membrane/metabolism , Bruch Membrane/pathology , Bruch Membrane/ultrastructure , Extracellular Space/metabolism , Integrases/metabolism , Intracellular Space/metabolism , Lipofuscin/metabolism , Melanosomes/metabolism , Melanosomes/ultrastructure , Mice , Mice, Inbred C57BL , Phagosomes/metabolism , Phagosomes/ultrastructure , Protein Transport , Retinal Pigment Epithelium/ultrastructure
14.
Traffic ; 13(10): 1351-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22780869

ABSTRACT

The obligate intracellular liver stage of the Plasmodium parasite represents a bottleneck in the parasite life cycle and remains a promising target for therapeutic intervention. During this stage, parasites undergo dramatic morphological changes and achieve one of the fastest replication rates among eukaryotic species. Nevertheless, relatively little is known about the parasite interactions with the host hepatocyte. Using immunofluorescence, live cell imaging and electron microscopy, we show that Plasmodium berghei parasites are surrounded by vesicles from the host late endocytic pathway. We found that these vesicles are acidic and contain the membrane markers Rab7a, CD63 and LAMP1. When host cell vesicle acidification was disrupted using ammonium chloride or Concanamycin A during the late liver stage of infection, parasite survival was not affected, but schizont size was significantly decreased. Furthermore, when the host cell endocytic pathway was loaded with BSA-gold, gold particles were found within the parasite cytoplasm, showing the transport of material from the host endocytic pathway toward the parasite interior. These observations reveal a novel Plasmodium-host interaction and suggest that vesicles from the host endolysosomal pathway could represent an important source of nutrients exploited by the fast-growing late liver stage parasites.


Subject(s)
Endocytosis , Hepatocytes/metabolism , Hepatocytes/parasitology , Host-Parasite Interactions , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Animals , Cytoplasmic Vesicles/chemistry , Female , Lysosomal Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Schizonts/growth & development , Tetraspanin 30/analysis , Tumor Cells, Cultured , rab GTP-Binding Proteins/analysis , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...