Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Annu Rev Biochem ; 86: 659-684, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28301744

ABSTRACT

The endoplasmic reticulum (ER) has a broad localization throughout the cell and forms direct physical contacts with all other classes of membranous organelles, including the plasma membrane (PM). A number of protein tethers that mediate these contacts have been identified, and study of these protein tethers has revealed a multiplicity of roles in cell physiology, including regulation of intracellular Ca2+ dynamics and signaling as well as control of lipid traffic and homeostasis. In this review, we discuss the cross talk between the ER and the PM mediated by direct contacts. We review factors that tether the two membranes, their properties, and their dynamics in response to the functional state of the cell. We focus in particular on the role of ER-PM contacts in nonvesicular lipid transport between the two bilayers mediated by lipid transfer proteins.


Subject(s)
Calcium/metabolism , Carrier Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , ORAI1 Protein/metabolism , Biological Transport , Calcium Signaling , Carrier Proteins/genetics , Cell Membrane/ultrastructure , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Gene Expression , Homeostasis , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein/genetics , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Synaptotagmins/genetics , Synaptotagmins/metabolism
3.
Cell ; 166(2): 408-423, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27419871

ABSTRACT

VAP (VAPA and VAPB) is an evolutionarily conserved endoplasmic reticulum (ER)-anchored protein that helps generate tethers between the ER and other membranes through which lipids are exchanged across adjacent bilayers. Here, we report that by regulating PI4P levels on endosomes, VAP affects WASH-dependent actin nucleation on these organelles and the function of the retromer, a protein coat responsible for endosome-to-Golgi traffic. VAP is recruited to retromer budding sites on endosomes via an interaction with the retromer SNX2 subunit. Cells lacking VAP accumulate high levels of PI4P, actin comets, and trans-Golgi proteins on endosomes. Such defects are mimicked by downregulation of OSBP, a VAP interactor and PI4P transporter that participates in VAP-dependent ER-endosomes tethers. These results reveal a role of PI4P in retromer-/WASH-dependent budding from endosomes. Collectively, our data show how the ER can control budding dynamics and association with the cytoskeleton of another membrane by direct contacts leading to bilayer lipid modifications.


Subject(s)
Endosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Vesicular Transport Proteins/metabolism , Actins/metabolism , Endoplasmic Reticulum/metabolism , Gene Knockout Techniques , HeLa Cells , Humans , Microfilament Proteins/metabolism , Receptors, Steroid/metabolism , Transcription Activator-Like Effector Nucleases , Vesicular Transport Proteins/genetics
4.
Cell ; 162(2): 425-440, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186194

ABSTRACT

Protein interactions form a network whose structure drives cellular function and whose organization informs biological inquiry. Using high-throughput affinity-purification mass spectrometry, we identify interacting partners for 2,594 human proteins in HEK293T cells. The resulting network (BioPlex) contains 23,744 interactions among 7,668 proteins with 86% previously undocumented. BioPlex accurately depicts known complexes, attaining 80%-100% coverage for most CORUM complexes. The network readily subdivides into communities that correspond to complexes or clusters of functionally related proteins. More generally, network architecture reflects cellular localization, biological process, and molecular function, enabling functional characterization of thousands of proteins. Network structure also reveals associations among thousands of protein domains, suggesting a basis for examining structurally related proteins. Finally, BioPlex, in combination with other approaches, can be used to reveal interactions of biological or clinical significance. For example, mutations in the membrane protein VAPB implicated in familial amyotrophic lateral sclerosis perturb a defined community of interactors.


Subject(s)
Protein Interaction Maps , Proteomics/methods , Amyotrophic Lateral Sclerosis/genetics , Humans , Mass Spectrometry , Protein Interaction Mapping , Proteins/chemistry , Proteins/isolation & purification , Proteins/metabolism
5.
Cell ; 153(7): 1494-509, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23791178

ABSTRACT

Most available information on endoplasmic reticulum (ER)-plasma membrane (PM) contacts in cells of higher eukaryotes concerns proteins implicated in the regulation of Ca(2+) entry. However, growing evidence suggests that such contacts play more general roles in cell physiology, pointing to the existence of additionally ubiquitously expressed ER-PM tethers. Here, we show that the three extended synaptotagmins (E-Syts) are ER proteins that participate in such tethering function via C2 domain-dependent interactions with the PM that require PI(4,5)P2 in the case of E-Syt2 and E-Syt3 and also elevation of cytosolic Ca(2+) in the case of E-Syt1. As they form heteromeric complexes, the E-Syts confer cytosolic Ca(2+) regulation to ER-PM contact formation. E-Syts-dependent contacts, however, are not required for store-operated Ca(2+) entry. Thus, the ER-PM tethering function of the E-Syts (tricalbins in yeast) mediates the formation of ER-PM contacts sites, which are functionally distinct from those mediated by STIM1 and Orai1.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Synaptotagmins/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/ultrastructure , HeLa Cells , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Synaptotagmins/chemistry , Synaptotagmins/genetics , Yeasts/cytology , Yeasts/metabolism
6.
Proc Natl Acad Sci U S A ; 121(17): e2318943121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38635628

ABSTRACT

Synaptojanin-1 (SJ1) is a major neuronal-enriched PI(4, 5)P2 4- and 5-phosphatase implicated in the shedding of endocytic factors during endocytosis. A mutation (R258Q) that impairs selectively its 4-phosphatase activity causes Parkinsonism in humans and neurological defects in mice (SJ1RQKI mice). Studies of these mice showed, besides an abnormal assembly state of endocytic factors at synapses, the presence of dystrophic nerve terminals selectively in a subset of nigro-striatal dopamine (DA)-ergic axons, suggesting a special lability of DA neurons to the impairment of SJ1 function. Here we have further investigated the impact of SJ1 on DA neurons using iPSC-derived SJ1 KO and SJ1RQKI DA neurons and their isogenic controls. In addition to the expected enhanced clustering of endocytic factors in nerve terminals, we observed in both SJ1 mutant neuronal lines increased cilia length. Further analysis of cilia of SJ1RQDA neurons revealed abnormal accumulation of the Ca2+ channel Cav1.3 and of ubiquitin chains, suggesting a defect in the clearing of ubiquitinated proteins at the ciliary base, where a focal concentration of SJ1 was observed. We suggest that SJ1 may contribute to the control of ciliary protein dynamics in DA neurons, with implications on cilia-mediated signaling.


Subject(s)
Induced Pluripotent Stem Cells , Nerve Tissue Proteins , Parkinson Disease , Parkinsonian Disorders , Humans , Mice , Animals , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Mutation
7.
Proc Natl Acad Sci U S A ; 120(43): e2309698120, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37844218

ABSTRACT

Mutations in Leucine-rich repeat kinase 2 (LRRK2) are responsible for late-onset autosomal dominant Parkinson's disease. LRRK2 has been implicated in a wide range of physiological processes including membrane repair in the endolysosomal system. Here, using cell-free systems, we report that purified LRRK2 directly binds acidic lipid bilayers with a preference for highly curved bilayers. While this binding is nucleotide independent, LRRK2 can also deform low-curvature liposomes into narrow tubules in a guanylnucleotide-dependent but Adenosine 5'-triphosphate-independent way. Moreover, assembly of LRRK2 into scaffolds at the surface of lipid tubules can constrict them. We suggest that an interplay between the membrane remodeling and signaling properties of LRRK2 may be key to its physiological function. LRRK2, via its kinase activity, may achieve its signaling role at sites where membrane remodeling occurs.


Subject(s)
Parkinson Disease , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation , Mutation
8.
Proc Natl Acad Sci U S A ; 119(42): e2203750119, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36215465

ABSTRACT

The spine apparatus is a specialized compartment of the neuronal smooth endoplasmic reticulum (ER) located in a subset of dendritic spines. It consists of stacks of ER cisterns that are interconnected by an unknown dense matrix and are continuous with each other and with the ER of the dendritic shaft. While this organelle was first observed over 60 y ago, its molecular organization remains a mystery. Here, we performed in vivo proximity proteomics to gain some insight into its molecular components. To do so, we used the only known spine apparatus-specific protein, synaptopodin, to target a biotinylating enzyme to this organelle. We validated the specific localization in dendritic spines of a small subset of proteins identified by this approach, and we further showed their colocalization with synaptopodin when expressed in nonneuronal cells. One such protein is Pdlim7, an actin binding protein not previously identified in spines. Pdlim7, which we found to interact with synaptopodin through multiple domains, also colocalizes with synaptopodin on the cisternal organelle, a peculiar stack of ER cisterns resembling the spine apparatus and found at axon initial segments of a subset of neurons. Moreover, Pdlim7 has an expression pattern similar to that of synaptopodin in the brain, highlighting a functional partnership between the two proteins. The components of the spine apparatus identified in this work will help elucidate mechanisms in the biogenesis and maintenance of this enigmatic structure with implications for the function of dendritic spines in physiology and disease.


Subject(s)
Dendritic Spines , Proteomics , Dendritic Spines/metabolism , Endoplasmic Reticulum/metabolism , Hippocampus/metabolism , Microfilament Proteins/metabolism
9.
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
10.
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
11.
Genet Med ; : 101166, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38767059

ABSTRACT

PURPOSE: The function of FAM177A1 and its relationship to human disease is largely unknown. Recent studies have demonstrated FAM177A1 to be a critical immune-associated gene. One previous case study has linked FAM177A1 to a neurodevelopmental disorder in four siblings. METHODS: We identified five individuals from three unrelated families with biallelic variants in FAM177A1. The physiological function of FAM177A1 was studied in a zebrafish model organism and human cell lines with loss-of-function variants similar to the affected cohort. RESULTS: These individuals share a characteristic phenotype defined by macrocephaly, global developmental delay, intellectual disability, seizures, behavioral abnormalities, hypotonia, and gait disturbance. We show that FAM177A1 localizes to the Golgi complex in mammalian and zebrafish cells. Intersection of the RNA-seq and metabolomic datasets from FAM177A1-deficient human fibroblasts and whole zebrafish larvae demonstrated dysregulation of pathways associated with apoptosis, inflammation, and negative regulation of cell proliferation. CONCLUSION: Our data sheds light on the emerging function of FAM177A1 and defines FAM177A1-related neurodevelopmental disorder as a new clinical entity.

12.
Nat Chem Biol ; 18(3): 313-320, 2022 03.
Article in English | MEDLINE | ID: mdl-34916620

ABSTRACT

Extended synaptotagmins (E-Syts) mediate lipid exchange between the endoplasmic reticulum (ER) and the plasma membrane (PM). Anchored on the ER, E-Syts bind the PM via an array of C2 domains in a Ca2+- and lipid-dependent manner, drawing the two membranes close to facilitate lipid exchange. How these C2 domains bind the PM and regulate the ER-PM distance is not well understood. Here, we applied optical tweezers to dissect PM binding by E-Syt1 and E-Syt2. We detected Ca2+- and lipid-dependent membrane-binding kinetics of both E-Syts and determined the binding energies and rates of individual C2 domains or pairs. We incorporated these parameters in a theoretical model to recapitulate salient features of E-Syt-mediated membrane contacts observed in vivo, including their equilibrium distances and probabilities. Our methods can be applied to study other proteins containing multiple membrane-binding domains linked by disordered polypeptides.


Subject(s)
Calcium , Optical Tweezers , Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Lipids/analysis
13.
Nat Rev Mol Cell Biol ; 13(2): 75-88, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22233676

ABSTRACT

Dynamin, the founding member of a family of dynamin-like proteins (DLPs) implicated in membrane remodelling, has a critical role in endocytic membrane fission events. The use of complementary approaches, including live-cell imaging, cell-free studies, X-ray crystallography and genetic studies in mice, has greatly advanced our understanding of the mechanisms by which dynamin acts, its essential roles in cell physiology and the specific function of different dynamin isoforms. In addition, several connections between dynamin and human disease have also emerged, highlighting specific contributions of this GTPase to the physiology of different tissues.


Subject(s)
Cell Membrane/physiology , Dynamins/physiology , Animals , Cell Membrane/metabolism , Dynamins/chemistry , Dynamins/genetics , Dynamins/metabolism , Endocytosis/genetics , Endocytosis/physiology , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/physiology , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Mammals/genetics , Mammals/metabolism , Membrane Fluidity/genetics , Mice , Models, Biological , Models, Molecular , Protein Conformation
14.
Cell ; 137(2): 191-6, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19379681

ABSTRACT

Membrane-shaping proteins of the BAR domain superfamily are determinants of organelle biogenesis, membrane trafficking, cell division, and cell migration. An upsurge of research now reveals new principles of BAR domain-mediated membrane remodeling, enhancing our understanding of membrane curvature-mediated information processing.


Subject(s)
Membrane Proteins/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cytoskeleton/metabolism , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Protein Structure, Tertiary
15.
Cell ; 136(6): 1110-21, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19303853

ABSTRACT

The recent identification of several novel endocytic compartments has challenged our current understanding of the topological and functional organization of the endocytic pathway. Using quantitative single vesicle imaging and acute manipulation of phosphoinositides we show that APPL endosomes, which participate in growth factor receptor trafficking and signaling, represent an early endocytic intermediate common to a subset of clathrin derived endocytic vesicles and macropinosomes. Most APPL endosomes are precursors of classical PI3P positive endosomes, and PI3P plays a critical role in promoting this conversion. Depletion of PI3P causes a striking reversion of Rab5 positive endosomes to the APPL stage, and results in enhanced growth factor signaling. These findings reveal a surprising plasticity of the early endocytic pathway. Importantly, PI3P functions as a switch to dynamically regulate maturation and signaling of APPL endosomes.


Subject(s)
Endosomes/metabolism , Phosphatidylinositols/metabolism , Animals , COS Cells , Chlorocebus aethiops , Clathrin-Coated Vesicles/metabolism , Endocytosis , Signal Transduction
16.
Nat Methods ; 17(2): 225-231, 2020 02.
Article in English | MEDLINE | ID: mdl-31907447

ABSTRACT

Combining the molecular specificity of fluorescent probes with three-dimensional imaging at nanoscale resolution is critical for investigating the spatial organization and interactions of cellular organelles and protein complexes. We present a 4Pi single-molecule switching super-resolution microscope that enables ratiometric multicolor imaging of mammalian cells at 5-10-nm localization precision in three dimensions using 'salvaged fluorescence'. Imaging two or three fluorophores simultaneously, we show fluorescence images that resolve the highly convoluted Golgi apparatus and the close contacts between the endoplasmic reticulum and the plasma membrane, structures that have traditionally been the imaging realm of electron microscopy. The salvaged fluorescence approach is equally applicable in most single-objective microscopes.


Subject(s)
Optical Imaging , Subcellular Fractions/metabolism , Animals , Humans , Organelles/metabolism
17.
Cell ; 132(5): 807-17, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18329367

ABSTRACT

BAR superfamily domains shape membranes through poorly understood mechanisms. We solved structures of F-BAR modules bound to flat and curved bilayers using electron (cryo)microscopy. We show that membrane tubules form when F-BARs polymerize into helical coats that are held together by lateral and tip-to-tip interactions. On gel-state membranes or after mutation of residues along the lateral interaction surface, F-BARs adsorb onto bilayers via surfaces other than their concave face. We conclude that membrane binding is separable from membrane bending, and that imposition of the module's concave surface forces fluid-phase bilayers to bend locally. Furthermore, exposure of the domain's lateral interaction surface through a change in orientation serves as the crucial trigger for assembly of the helical coat and propagation of bilayer bending. The geometric constraints and sequential assembly of the helical lattice explain how F-BAR and classical BAR domains segregate into distinct microdomains, and provide insight into the spatial regulation of membrane invagination.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Animals , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/ultrastructure , Chlorocebus aethiops , Cryoelectron Microscopy , Dynamins/metabolism , Fatty Acid-Binding Proteins , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Liposomes/chemistry , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Minor Histocompatibility Antigens , Models, Biological , Models, Molecular , Protein Structure, Tertiary , Transfection
18.
Proc Natl Acad Sci U S A ; 117(22): 12428-12434, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32424101

ABSTRACT

Numerous genes whose mutations cause, or increase the risk of, Parkinson's disease (PD) have been identified. An inactivating mutation (R258Q) in the Sac inositol phosphatase domain of synaptojanin 1 (SJ1/PARK20), a phosphoinositide phosphatase implicated in synaptic vesicle recycling, results in PD. The gene encoding Sac2/INPP5F, another Sac domain-containing protein, is located within a PD risk locus identified by genome-wide association studies. Knock-In mice carrying the SJ1 patient mutation (SJ1RQKI) exhibit PD features, while Sac2 knockout mice (Sac2KO) do not have obvious neurologic defects. We report a "synthetic" effect of the SJ1 mutation and the KO of Sac2 in mice. Most mice with both mutations died perinatally. The occasional survivors had stunted growth, died within 3 wk, and showed abnormalities of striatal dopaminergic nerve terminals at an earlier stage than SJ1RQKI mice. The abnormal accumulation of endocytic factors observed at synapses of cultured SJ1RQKI neurons was more severe in double-mutant neurons. Our results suggest that SJ1 and Sac2 have partially overlapping functions and are consistent with a potential role of Sac2 as a PD risk gene.


Subject(s)
Inositol Polyphosphate 5-Phosphatases/genetics , Parkinson Disease/enzymology , Animals , Dopamine/metabolism , Genome-Wide Association Study , Humans , Inositol Polyphosphate 5-Phosphatases/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Endings/metabolism , Parkinson Disease/genetics , Phenotype , Synapses/metabolism
19.
EMBO J ; 37(2): 219-234, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29222176

ABSTRACT

The extended synaptotagmins (E-Syts) are endoplasmic reticulum (ER) proteins that bind the plasma membrane (PM) via C2 domains and transport lipids between them via SMP domains. E-Syt1 tethers and transports lipids in a Ca2+-dependent manner, but the role of Ca2+ in this regulation is unclear. Of the five C2 domains of E-Syt1, only C2A and C2C contain Ca2+-binding sites. Using liposome-based assays, we show that Ca2+ binding to C2C promotes E-Syt1-mediated membrane tethering by releasing an inhibition that prevents C2E from interacting with PI(4,5)P2-rich membranes, as previously suggested by studies in semi-permeabilized cells. Importantly, Ca2+ binding to C2A enables lipid transport by releasing a charge-based autoinhibitory interaction between this domain and the SMP domain. Supporting these results, E-Syt1 constructs defective in Ca2+ binding in either C2A or C2C failed to rescue two defects in PM lipid homeostasis observed in E-Syts KO cells, delayed diacylglycerol clearance from the PM and impaired Ca2+-triggered phosphatidylserine scrambling. Thus, a main effect of Ca2+ on E-Syt1 is to reverse an autoinhibited state and to couple membrane tethering with lipid transport.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Lipid Metabolism/physiology , Synaptotagmins/metabolism , Biological Transport, Active/physiology , Cell Line , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , Humans , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Domains , Synaptotagmins/genetics
20.
Brain ; 144(12): 3597-3610, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34415310

ABSTRACT

Phosphatidylinositol 4-kinase IIIα (PI4KIIIα/PI4KA/OMIM:600286) is a lipid kinase generating phosphatidylinositol 4-phosphate (PI4P), a membrane phospholipid with critical roles in the physiology of multiple cell types. PI4KIIIα's role in PI4P generation requires its assembly into a heterotetrameric complex with EFR3, TTC7 and FAM126. Sequence alterations in two of these molecular partners, TTC7 (encoded by TTC7A or TCC7B) and FAM126, have been associated with a heterogeneous group of either neurological (FAM126A) or intestinal and immunological (TTC7A) conditions. Here we show that biallelic PI4KA sequence alterations in humans are associated with neurological disease, in particular hypomyelinating leukodystrophy. In addition, affected individuals may present with inflammatory bowel disease, multiple intestinal atresia and combined immunodeficiency. Our cellular, biochemical and structural modelling studies indicate that PI4KA-associated phenotypical outcomes probably stem from impairment of PI4KIIIα-TTC7-FAM126's organ-specific functions, due to defective catalytic activity or altered intra-complex functional interactions. Together, these data define PI4KA gene alteration as a cause of a variable phenotypical spectrum and provide fundamental new insight into the combinatorial biology of the PI4KIIIα-FAM126-TTC7-EFR3 molecular complex.


Subject(s)
Hereditary Central Nervous System Demyelinating Diseases/genetics , Intestinal Atresia/genetics , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Primary Immunodeficiency Diseases/genetics , Female , Humans , Male , Pedigree , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL