Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 22(5): e3002639, 2024 May.
Article in English | MEDLINE | ID: mdl-38820535

ABSTRACT

Vesicular trafficking, including secretion and endocytosis, plays fundamental roles in the unique biology of Plasmodium falciparum blood-stage parasites. Endocytosis of host cell cytosol (HCC) provides nutrients and room for parasite growth and is critical for the action of antimalarial drugs and parasite drug resistance. Previous work showed that PfVPS45 functions in endosomal transport of HCC to the parasite's food vacuole, raising the possibility that malaria parasites possess a canonical endolysosomal system. However, the seeming absence of VPS45-typical functional interactors such as rabenosyn 5 (Rbsn5) and the repurposing of Rab5 isoforms and other endolysosomal proteins for secretion in apicomplexans question this idea. Here, we identified a parasite Rbsn5-like protein and show that it functions with VPS45 in the endosomal transport of HCC. We also show that PfRab5b but not PfRab5a is involved in the same process. Inactivation of PfRbsn5L resulted in PI3P and PfRab5b decorated HCC-filled vesicles, typical for endosomal compartments. Overall, this indicates that despite the low sequence conservation of PfRbsn5L and the unusual N-terminal modification of PfRab5b, principles of endosomal transport in malaria parasite are similar to that of model organisms. Using a conditional double protein inactivation system, we further provide evidence that the PfKelch13 compartment, an unusual apicomplexa-specific endocytosis structure at the parasite plasma membrane, is connected upstream of the Rbsn5L/VPS45/Rab5b-dependent endosomal route. Altogether, this work indicates that HCC uptake consists of a highly parasite-specific part that feeds endocytosed material into an endosomal system containing more canonical elements, leading to the delivery of HCC to the food vacuole.


Subject(s)
Cytosol , Endosomes , Plasmodium falciparum , Protozoan Proteins , rab5 GTP-Binding Proteins , rab5 GTP-Binding Proteins/metabolism , Endosomes/metabolism , Cytosol/metabolism , Plasmodium falciparum/metabolism , Plasmodium falciparum/genetics , Humans , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Endocytosis , Malaria, Falciparum/parasitology , Malaria, Falciparum/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Animals , Host-Parasite Interactions , Vacuoles/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism , Protein Transport
2.
Biomedicines ; 11(10)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37893031

ABSTRACT

Sensorineural age-related hearing loss affects a large proportion of the elderly population, and has both environmental and genetic causes. Notwithstanding increasing interest in this debilitating condition, the genetic risk factors remain largely unknown. Here, we report the case of two sisters affected by isolated profound sensorineural hearing loss after the age of seventy. Genomic DNA sequencing revealed that the siblings shared two monoallelic variants in two genes linked to Usher Syndrome (USH genes), a recessive disorder of the ear and the retina: a rare pathogenic truncating variant in USH1G and a previously unreported missense variant in ADGRV1. Structure predictions suggest a negative effect on protein stability of the latter variant, allowing its classification as likely pathogenic according to American College of Medical Genetics criteria. Thus, the presence in heterozygosis of two recessive alleles, which each cause syndromic deafness, may underlie digenic inheritance of the age-related non-syndromic hearing loss of the siblings, a hypothesis that is strengthened by the knowledge that the two genes are integrated in the same functional network, which underlies stereocilium development and organization. These results enlarge the spectrum and complexity of the phenotypic consequences of USH gene mutations beyond the simple Mendelian inheritance of classical Usher syndrome.

3.
Annu Rev Cell Dev Biol ; 39: 409-434, 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37406299

ABSTRACT

The life of eukaryotic cells requires the transport of lipids between membranes, which are separated by the aqueous environment of the cytosol. Vesicle-mediated traffic along the secretory and endocytic pathways and lipid transfer proteins (LTPs) cooperate in this transport. Until recently, known LTPs were shown to carry one or a few lipids at a time and were thought to mediate transport by shuttle-like mechanisms. Over the last few years, a new family of LTPs has been discovered that is defined by a repeating ß-groove (RBG) rod-like structure with a hydrophobic channel running along their entire length. This structure and the localization of these proteins at membrane contact sites suggest a bridge-like mechanism of lipid transport. Mutations in some of these proteins result in neurodegenerative and developmental disorders. Here we review the known properties and well-established or putative physiological roles of these proteins, and we highlight the many questions that remain open about their functions.


Subject(s)
Carrier Proteins , Proteins , Carrier Proteins/chemistry , Proteins/metabolism , Biological Transport/genetics , Cell Membrane/metabolism , Lipids/chemistry
4.
Article in English | MEDLINE | ID: mdl-36123033

ABSTRACT

The Endoplasmic Reticulum (ER) is an endomembrane system that plays a multiplicity of roles in cell physiology and populates even the most distal cell compartments, including dendritic tips and axon terminals of neurons. Some of its functions are achieved by a cross talk with other intracellular membranous organelles and with the plasma membrane at membrane contacts sites (MCSs). As the ER synthesizes most membrane lipids, lipid exchanges mediated by lipid transfer proteins at MCSs are a particularly important aspect of this cross talk, which synergizes with the cross talk mediated by vesicular transport. Several mutations of genes that encode proteins localized at ER MCSs result in familial neurodegenerative diseases, emphasizing the importance of the normal lipid traffic within cells for a healthy brain. Here, we provide an overview of such diseases, with a specific focus on proteins that directly or indirectly impact lipid transport.


Subject(s)
Endoplasmic Reticulum , Lipids , Endoplasmic Reticulum/metabolism , Biological Transport , Cell Membrane/metabolism , Lipid Metabolism
5.
Proc Natl Acad Sci U S A ; 119(35): e2205425119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994651

ABSTRACT

Chorea-acanthocytosis (ChAc) and McLeod syndrome are diseases with shared clinical manifestations caused by mutations in VPS13A and XK, respectively. Key features of these conditions are the degeneration of caudate neurons and the presence of abnormally shaped erythrocytes. XK belongs to a family of plasma membrane (PM) lipid scramblases whose action results in exposure of PtdSer at the cell surface. VPS13A is an endoplasmic reticulum (ER)-anchored lipid transfer protein with a putative role in the transport of lipids at contacts of the ER with other membranes. Recently VPS13A and XK were reported to interact by still unknown mechanisms. So far, however, there is no evidence for a colocalization of the two proteins at contacts of the ER with the PM, where XK resides, as VPS13A was shown to be localized at contacts between the ER and either mitochondria or lipid droplets. Here we show that VPS13A can also localize at ER-PM contacts via the binding of its PH domain to a cytosolic loop of XK, that such interaction is regulated by an intramolecular interaction within XK, and that both VPS13A and XK are highly expressed in the caudate neurons. Binding of the PH domain of VPS13A to XK is competitive with its binding to intracellular membranes that mediate other tethering functions of VPS13A. Our findings support a model according to which VPS13A-dependent lipid transfer between the ER and the PM is coupled to lipid scrambling within the PM. They raise the possibility that defective cell surface exposure of PtdSer may be responsible for neurodegeneration.


Subject(s)
Carrier Proteins , Cell Membrane , Lipids , Vesicular Transport Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Humans , Neuroacanthocytosis/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 119(29): e2203769119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858323

ABSTRACT

VPS13 is a eukaryotic lipid transport protein localized at membrane contact sites. Previous studies suggested that it may transfer lipids between adjacent bilayers by a bridge-like mechanism. Direct evidence for this hypothesis from a full-length structure and from electron microscopy (EM) studies in situ is still missing, however. Here, we have capitalized on AlphaFold predictions to complement the structural information already available about VPS13 and to generate a full-length model of human VPS13C, the Parkinson's disease-linked VPS13 paralog localized at contacts between the endoplasmic reticulum (ER) and endo/lysosomes. Such a model predicts an ∼30-nm rod with a hydrophobic groove that extends throughout its length. We further investigated whether such a structure can be observed in situ at ER-endo/lysosome contacts. To this aim, we combined genetic approaches with cryo-focused ion beam (cryo-FIB) milling and cryo-electron tomography (cryo-ET) to examine HeLa cells overexpressing this protein (either full length or with an internal truncation) along with VAP, its anchoring binding partner at the ER. Using these methods, we identified rod-like densities that span the space separating the two adjacent membranes and that match the predicted structures of either full-length VPS13C or its shorter truncated mutant, thus providing in situ evidence for a bridge model of VPS13 in lipid transport.


Subject(s)
Endoplasmic Reticulum , Lipid Metabolism , Proteins , ATP-Binding Cassette Transporters , Bacterial Outer Membrane Proteins , Biological Transport , Cell Membrane/chemistry , Cryoelectron Microscopy , Endoplasmic Reticulum/chemistry , HeLa Cells , Humans , Lysosomes/chemistry , Proteins/chemistry
8.
J Cell Biol ; 220(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33891013

ABSTRACT

Mitochondria, which are excluded from the secretory pathway, depend on lipid transport proteins for their lipid supply from the ER, where most lipids are synthesized. In yeast, the outer mitochondrial membrane GTPase Gem1 is an accessory factor of ERMES, an ER-mitochondria tethering complex that contains lipid transport domains and that functions, partially redundantly with Vps13, in lipid transfer between the two organelles. In metazoa, where VPS13, but not ERMES, is present, the Gem1 orthologue Miro was linked to mitochondrial dynamics but not to lipid transport. Here we show that Miro, including its peroxisome-enriched splice variant, recruits the lipid transport protein VPS13D, which in turn binds the ER in a VAP-dependent way and thus could provide a lipid conduit between the ER and mitochondria. These findings reveal a so far missing link between function(s) of Gem1/Miro in yeast and higher eukaryotes, where Miro is a Parkin substrate, with potential implications for Parkinson's disease pathogenesis.


Subject(s)
Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , Proteins/metabolism , Animals , Biological Transport/physiology , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Eukaryota/metabolism , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , Mitochondrial Dynamics/physiology , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Mol Psychiatry ; 26(7): 2872-2885, 2021 07.
Article in English | MEDLINE | ID: mdl-33742167

ABSTRACT

Among the hallmarks of major depressive disorders (MDD) are molecular, functional, and morphological impairments in the hippocampus. Recent studies suggested a key role for hippocampal GABAergic interneurons both in depression and in the response to its treatments. These interneurons highly express the chromatin-remodeler SMARCA3 which mediates the response to chronic antidepressants in an unknown mechanism. Using cell-type-specific molecular and physiological approaches, we report that SMARCA3 mediates the glutamatergic signaling in interneurons by repressing the expression of the neuronal protein, Neurensin-2. This vesicular protein associates with endosomes and postsynaptic proteins and is highly and selectively expressed in subpopulations of GABAergic interneurons. Upregulation of Neurensin-2 in the hippocampus either by stress, viral overexpression, or by SMARCA3 deletion, results in depressive-like behaviors. In contrast, the deletion of Neurensin-2 confers resilience to stress and induces AMPA receptor localization to synapses. This pathway which bidirectionally affects emotional behavior could be involved in neuropsychiatric disorders, and suggests novel therapeutic approaches.


Subject(s)
Depressive Disorder, Major , Hippocampus , Humans , Interneurons , Neurons , Synapses
10.
Elife ; 92020 11 11.
Article in English | MEDLINE | ID: mdl-33174843

ABSTRACT

Light-inducible dimerization protein modules enable precise temporal and spatial control of biological processes in non-invasive fashion. Among them, Magnets are small modules engineered from the Neurospora crassa photoreceptor Vivid by orthogonalizing the homodimerization interface into complementary heterodimers. Both Magnets components, which are well-tolerated as protein fusion partners, are photoreceptors requiring simultaneous photoactivation to interact, enabling high spatiotemporal confinement of dimerization with a single excitation wavelength. However, Magnets require concatemerization for efficient responses and cell preincubation at 28°C to be functional. Here we overcome these limitations by engineering an optimized Magnets pair requiring neither concatemerization nor low temperature preincubation. We validated these 'enhanced' Magnets (eMags) by using them to rapidly and reversibly recruit proteins to subcellular organelles, to induce organelle contacts, and to reconstitute OSBP-VAP ER-Golgi tethering implicated in phosphatidylinositol-4-phosphate transport and metabolism. eMags represent a very effective tool to optogenetically manipulate physiological processes over whole cells or in small subcellular volumes.


The cell relies on direct interactions among proteins and compartments called organelles to stay alive. Manipulating these interactions allows researchers to control a wide variety of cell behaviors. A system called 'Magnets' uses light to trigger interactions between proteins. Magnets uses a segment of a protein called Vivid from a common bread mold that responds to light. When light shines on two of these segments, it causes them to bind together, in a process known as dimerization. In the Magnets system, Vivid segments are attached to specific proteins or organelles. By using light, researchers can force their target molecules to come together and trigger signals that can change cell behavior. However, the Magnets system has limitations: its stability and low efficiency mean that the cells need to be kept at low temperatures and that several copies of Vivid are needed. These conditions can interfere with the activity of the target proteins. To expand the technique, Benedetti et al. added mutations to make the Vivid protein more similar to proteins found in fungi that thrive at temperatures around 50°C. These changes meant that the enhanced system could work at body temperature in mammals. Further mutations at the interface between the two Vivid segments improved the efficiency of dimerization. This enhanced version was put to the test in different applications, including delivering proteins to different organelles and bringing organelles together. The enhanced Magnets system should enable researchers to control a greater variety of signaling events in the cell. In addition, the methodology established for improving the efficiency of the Magnets system could be useful to researchers working on other proteins.


Subject(s)
Biological Transport , Fungal Proteins/radiation effects , Light , Optogenetics , Organelles/metabolism , Protein Engineering , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Fungal Proteins/genetics , Fungal Proteins/metabolism , HeLa Cells , Humans , Kinetics , Lipid Metabolism , Mice, Inbred C57BL , Organelles/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Multimerization , Protein Stability , Protein Transport
11.
Proc Natl Acad Sci U S A ; 116(45): 22619-22623, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31636202

ABSTRACT

Contacts between the endoplasmic reticulum (ER) and other membranes are hot spots for protein-mediated lipid transport between the 2 adjacent bilayers. Compiling a molecular inventory of lipid transport proteins present at these sites is a premise to the elucidation of their function. Here we show that PDZD8, an intrinsic membrane protein of the ER with a lipid transport module of the SMP domain family, concentrates at contacts between the ER and late endosomes/lysosomes, where it interacts with GTP-Rab7. These findings suggest that PDZD8 may cooperate with other proteins that function at the ER-endo/lysosome interface in coordinating endocytic flow with lipid transport between endocytic membranes and the ER.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lysosomes/metabolism , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum/genetics , Endosomes/genetics , Humans , Lysosomes/genetics , Protein Binding , Protein Domains , Protein Transport , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
12.
Proc Natl Acad Sci U S A ; 116(12): 5775-5784, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30819882

ABSTRACT

Close appositions between the endoplasmic reticulum (ER) and the plasma membrane (PM) are a general feature of all cells and are abundant in neurons. A function of these appositions is lipid transport between the two adjacent bilayers via tethering proteins that also contain lipid transport modules. However, little is known about the properties and dynamics of these proteins in neurons. Here we focused on TMEM24/C2CD2L, an ER-localized SMP domain containing phospholipid transporter expressed at high levels in the brain, previously shown to be a component of ER-PM contacts in pancreatic ß-cells. TMEM24 is enriched in neurons versus glial cells and its levels increase in parallel with neuronal differentiation. It populates ER-PM contacts in resting neurons, but elevations of cytosolic Ca2+ mediated by experimental manipulations or spontaneous activity induce its transient redistribution throughout the entire ER. Dissociation of TMEM24 from the plasma membrane is mediated by phosphorylation of an array of sites in the C-terminal region of the protein. These sites are only partially conserved in C2CD2, the paralogue of TMEM24 primarily expressed in nonneuronal tissues, which correspondingly display a much lower sensitivity to Ca2+ elevations. ER-PM contacts in neurons are also sites where Kv2 (the major delayed rectifier K+ channels in brain) and other PM and ER ion channels are concentrated, raising the possibility of a regulatory feedback mechanism between neuronal excitability and lipid exchange between the ER and the PM.


Subject(s)
Calcium Signaling/physiology , Membrane Proteins/metabolism , Neurons/physiology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Humans , Lipids , Mammals/metabolism , Membrane Proteins/physiology , Mice , Neurons/metabolism , Phospholipids/metabolism , Phosphorylation , Primary Cell Culture , Synaptotagmins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...