Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell ; 183(6): 1520-1535.e14, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33157038

ABSTRACT

ß-Coronaviruses are a family of positive-strand enveloped RNA viruses that includes the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Much is known regarding their cellular entry and replication pathways, but their mode of egress remains uncertain. Using imaging methodologies and virus-specific reporters, we demonstrate that ß-coronaviruses utilize lysosomal trafficking for egress rather than the biosynthetic secretory pathway more commonly used by other enveloped viruses. This unconventional egress is regulated by the Arf-like small GTPase Arl8b and can be blocked by the Rab7 GTPase competitive inhibitor CID1067700. Such non-lytic release of ß-coronaviruses results in lysosome deacidification, inactivation of lysosomal degradation enzymes, and disruption of antigen presentation pathways. ß-Coronavirus-induced exploitation of lysosomal organelles for egress provides insights into the cellular and immunological abnormalities observed in patients and suggests new therapeutic modalities.


Subject(s)
COVID-19/metabolism , SARS-CoV-2/metabolism , Secretory Pathway , Virus Release , ADP-Ribosylation Factors/metabolism , Animals , COVID-19/pathology , Female , HeLa Cells , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Lysosomes , Mice , Thiourea/analogs & derivatives , Thiourea/pharmacology , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , COVID-19 Drug Treatment
2.
EMBO J ; 40(20): e107766, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34516001

ABSTRACT

The Golgi apparatus, the main glycosylation station of the cell, consists of a stack of discontinuous cisternae. Glycosylation enzymes are usually concentrated in one or two specific cisternae along the cis-trans axis of the organelle. How such compartmentalized localization of enzymes is achieved and how it contributes to glycosylation are not clear. Here, we show that the Golgi matrix protein GRASP55 directs the compartmentalized localization of key enzymes involved in glycosphingolipid (GSL) biosynthesis. GRASP55 binds to these enzymes and prevents their entry into COPI-based retrograde transport vesicles, thus concentrating them in the trans-Golgi. In genome-edited cells lacking GRASP55, or in cells expressing mutant enzymes without GRASP55 binding sites, these enzymes relocate to the cis-Golgi, which affects glycosphingolipid biosynthesis by changing flux across metabolic branch points. These findings reveal a mechanism by which a matrix protein regulates polarized localization of glycosylation enzymes in the Golgi and controls competition in glycan biosynthesis.


Subject(s)
Glycosphingolipids/metabolism , Golgi Apparatus/metabolism , Golgi Matrix Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Brefeldin A/pharmacology , Ceramides/metabolism , Cholera Toxin/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression , Glycosylation/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Golgi Matrix Proteins/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Shiga Toxin/pharmacology
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35173048

ABSTRACT

G protein-coupled receptors (GPCRs) play crucial roles in numerous physiological and pathological processes. Mutations in GPCRs that result in loss of function or alterations in signaling can lead to inherited or acquired diseases. Herein, studying prokineticin receptor 2 (PROKR2), we initially identify distinct interactomes for wild-type (WT) versus a mutant (P290S) PROKR2 that causes hypogonadotropic hypogonadism. We then find that both the WT and mutant PROKR2 are targeted for endoplasmic reticulum (ER)-associated degradation, but the mutant is degraded to a greater extent. Further analysis revealed that both forms can also leave the ER to reach the Golgi. However, whereas most of the WT is further transported to the cell surface, most of the mutant is retrieved to the ER. Thus, the post-ER itinerary plays an important role in distinguishing the ultimate fate of the WT versus the mutant. We have further discovered that this post-ER itinerary reduces ER stress induced by the mutant PROKR2. Moreover, we extend the core findings to another model GPCR. Our findings advance the understanding of disease pathogenesis induced by a mutation at a key residue that is conserved across many GPCRs and thus contributes to a fundamental understanding of the diverse mechanisms used by cellular quality control to accommodate misfolded proteins.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Proteostasis/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Hypogonadism/metabolism , Mutation, Missense/genetics , Protein Transport/genetics , Protein Transport/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Signal Transduction
4.
Nat Rev Mol Cell Biol ; 13(5): 323-8, 2012 04 13.
Article in English | MEDLINE | ID: mdl-22498832

ABSTRACT

Endocytic recycling returns proteins to the plasma membrane in many physiological contexts. Studies of these events have helped to elucidate fundamental mechanisms that underlie recycling. Recycling was for some time considered to be the exception to a general mechanism of active cargo sorting in multiple intracellular pathways. In recent years, studies have begun to reconcile this seeming disparity and also suggest explanations for why early recycling studies did not detect active sorting. Further articulation of this emerging trend has far-reaching implications for a deeper understanding of many physiological and pathological events that require recycling.


Subject(s)
Endocytosis , Endosomes/metabolism , Biological Transport, Active , Cell Polarity , Humans , Protein Sorting Signals , Protein Transport , Receptors, Transferrin/metabolism , Vesicular Transport Proteins/metabolism
5.
Nature ; 561(7722): 263-267, 2018 09.
Article in English | MEDLINE | ID: mdl-30209366

ABSTRACT

Starvation poses a fundamental challenge to cell survival. Whereas the role of autophagy in promoting energy homeostasis in this setting has been extensively characterized1, other mechanisms are less well understood. Here we reveal that glyceraldehyde 3-phosphate dehydrogenase (GAPDH) inhibits coat protein I (COPI) transport by targeting a GTPase-activating protein (GAP) towards ADP-ribosylation factor 1 (ARF1) to suppress COPI vesicle fission. GAPDH inhibits multiple other transport pathways, also by targeting ARF GAPs. Further characterization suggests that this broad inhibition is activated by the cell during starvation to reduce energy consumption. These findings reveal a remarkable level of coordination among the intracellular transport pathways that underlies a critical mechanism of cellular energy homeostasis.


Subject(s)
Energy Metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Homeostasis , Adenylate Kinase/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Autophagy , COP-Coated Vesicles/metabolism , Cell Line , Chlorocebus aethiops , Cricetulus , Fibroblasts , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/chemistry , Humans , Mice , Phosphorylation , Ribonucleotides/metabolism , Starvation
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33658379

ABSTRACT

The sorting nexin (SNX) family of proteins deform the membrane to generate transport carriers in endosomal pathways. Here, we elucidate how a prototypic member, SNX1, acts in this process. Performing cryoelectron microscopy, we find that SNX1 assembles into a protein lattice that consists of helical rows of SNX1 dimers wrapped around tubular membranes in a crosslinked fashion. We also visualize the details of this structure, which provides a molecular understanding of how various parts of SNX1 contribute to its ability to deform the membrane. Moreover, we have compared the SNX1 structure with a previously elucidated structure of an endosomal coat complex formed by retromer coupled to a SNX, which reveals how the molecular organization of the SNX in this coat complex is affected by retromer. The comparison also suggests insight into intermediary stages of assembly that results in the formation of the retromer-SNX coat complex on the membrane.


Subject(s)
Cell Membrane/metabolism , Protein Multimerization , Sorting Nexins/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Mice , Protein Structure, Quaternary , Sorting Nexins/chemistry , Sorting Nexins/ultrastructure
7.
Am J Respir Cell Mol Biol ; 65(5): 532-543, 2021 11.
Article in English | MEDLINE | ID: mdl-34166600

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a common, complex disease and a major cause of morbidity and mortality. Although multiple genetic determinants of COPD have been implicated by genome-wide association studies (GWASs), the pathophysiological significance of these associations remains largely unknown. From a COPD protein-protein interaction network module, we selected a network path between two COPD GWAS genes for validation studies: FAM13A (family with sequence similarity 13 member A)-AP3D1-CTGF- TGFß2. We find that TGFß2, FAM13A, and AP3D1 (but not CTGF) form a cellular protein complex. Functional characterization suggests that this complex mediates the secretion of TGFß2 through an AP-3 (adaptor protein 3)-dependent pathway, with FAM13A acting as a negative regulator by targeting a late stage of this transport that involves the dissociation of coat-cargo interaction. Moreover, we find that TGFß2 is a transmembrane protein that engages the AP-3 complex for delivery to the late endosomal compartments for subsequent secretion through exosomes. These results identify a pathophysiological context that unifies the biological network role of two COPD GWAS proteins and reveal novel mechanisms of cargo transport through an intracellular pathway.


Subject(s)
Adaptor Protein Complex 3/metabolism , Adaptor Protein Complex delta Subunits/metabolism , GTPase-Activating Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Transforming Growth Factor beta2/metabolism , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex delta Subunits/genetics , Cell Line , Exosomes/metabolism , GTPase-Activating Proteins/genetics , Genome-Wide Association Study , HEK293 Cells , Humans , Protein Interaction Maps/genetics , Protein Transport , Reproducibility of Results , Transforming Growth Factor beta2/genetics
8.
Nat Rev Mol Cell Biol ; 10(5): 360-4, 2009 05.
Article in English | MEDLINE | ID: mdl-19293819

ABSTRACT

The coat protein I (COPI) complex is considered to be one of the best-characterized coat complexes. Studies on how it functions in vesicle formation have provided seminal contributions to the general paradigm in vesicular transport that the ADP-ribosylation factor (ARF) small GTPases are key regulators of coat complexes. Here, we discuss emerging evidence that suggests the need to revise some long-held views on how COPI vesicle formation is achieved.


Subject(s)
Coat Protein Complex I/physiology , Coated Vesicles/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Coat Protein Complex I/metabolism , Humans
9.
Nature ; 521(7553): 529-32, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25945738

ABSTRACT

The Golgi complex has a central role in the intracellular sorting of secretory proteins. Anterograde transport through the Golgi has been explained by the movement of Golgi cisternae, known as cisternal maturation. Because this explanation is now appreciated to be incomplete, interest has developed in understanding tubules that connect the Golgi cisternae. Here we show that the coat protein I (COPI) complex sorts anterograde cargoes into these tubules in human cells. Moreover, the small GTPase CDC42 regulates bidirectional Golgi transport by targeting the dual functions of COPI in cargo sorting and carrier formation. CDC42 also directly imparts membrane curvature to promote COPI tubule formation. Our findings further reveal that COPI tubular transport complements cisternal maturation in explaining how anterograde Golgi transport is achieved, and that bidirectional COPI transport is modulated by environmental cues through CDC42.


Subject(s)
Coat Protein Complex I/metabolism , Golgi Apparatus/metabolism , cdc42 GTP-Binding Protein/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Coatomer Protein/metabolism , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Protein Transport , Receptors, LDL/metabolism , Viral Envelope Proteins/metabolism
10.
PLoS Comput Biol ; 15(7): e1007081, 2019 07.
Article in English | MEDLINE | ID: mdl-31291238

ABSTRACT

Studies on the Bin-Amphiphysin-Rvs (BAR) domain have advanced a fundamental understanding of how proteins deform membrane. We previously showed that a BAR domain in tandem with a Pleckstrin Homology (PH domain) underlies the assembly of ACAP1 (Arfgap with Coil-coil, Ankryin repeat, and PH domain I) into an unusual lattice structure that also uncovers a new paradigm for how a BAR protein deforms membrane. Here, we initially pursued computation-based refinement of the ACAP1 lattice to identify its critical protein contacts. Simulation studies then revealed how ACAP1, which dimerizes into a symmetrical structure in solution, is recruited asymmetrically to the membrane through dynamic behavior. We also pursued electron microscopy (EM)-based structural studies, which shed further insight into the dynamic nature of the ACAP1 lattice assembly. As ACAP1 is an unconventional BAR protein, our findings broaden the understanding of the mechanistic spectrum by which proteins assemble into higher-ordered structures to achieve membrane deformation.


Subject(s)
GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , Cell Membrane/metabolism , Dimerization , GTPase-Activating Proteins/chemistry , Humans , Pleckstrin Homology Domains , Protein Conformation
11.
J Cell Sci ; 126(Pt 23): 5313-6, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24293329

ABSTRACT

A FASEB Summer Research Conference entitled 'Arf and Rab family G proteins' was held in July 2013 at Snowmass Village, Snowmass, Colorado. Arfs and Rabs are two families of GTPases that control membrane trafficking in eukaryotic cells, and increasing evidence indicates that their functions are tightly coordinated. Because many workers in this field have focused on only one family, this meeting was designed to integrate our understanding of the two families. The conference was organized by Elizabeth Sztul (University of Alabama, Birmingham, USA) and Jim Casanova (University of Virginia, Charlottesville, USA), and provided an opportunity for approximately 90 scientists to communicate their work and discuss future directions for the field. The talks highlighted the structural, functional and regulatory properties of Arf and Rab GTPases and the need to develop coordinated approaches to investigate them. Here, we present the major themes that emerged from the meeting.


Subject(s)
ADP-Ribosylation Factors/genetics , rab GTP-Binding Proteins/genetics , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/metabolism , Animals , Gene Expression Regulation , Humans , Signal Transduction , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 109(52): 21354-9, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23223633

ABSTRACT

Intracellular recycling pathways play critical roles in internalizing membrane and fluid phase cargo and in balancing the inflow and outflow of membrane and cell surface molecules. To identify proteins involved in the regulation of endocytic recycling, we used an shRNA trafficking library and screened for changes in the surface expression of CD1a antigen-presenting molecules that follow an endocytic recycling route. We found that silencing of the ADP-ribosylation factor (Arf)-like small GTPase Arl13b led to a decrease in CD1a surface expression, diminished CD1a function, and delayed CD1a recycling, suggesting that Arl13b is involved in the regulation of endocytic recycling traffic. Arl13b appears to be required for the major route of endocytic trafficking, causing clustering of early endosomes and leading to the accumulation of endocytic cargo. Moreover, Arl13b colocalized with markers of the endocytic recycling pathway followed by CD1a, namely Arf6 and Rab22a. We also detected an interaction between Arl13b and the actin cytoskeleton. Arl13b was previously implicated in cilia formation and function. Our present results indicate a previously unidentified role for Arl13b in endocytic recycling traffic and suggest a link between Arl13b function and the actin cytoskeleton.


Subject(s)
ADP-Ribosylation Factors/metabolism , Endocytosis , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/chemistry , Actin Cytoskeleton/metabolism , Antigens, CD1/metabolism , Cell Membrane/metabolism , Cluster Analysis , Endosomes/metabolism , Gene Silencing , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Transferrin/metabolism , rab GTP-Binding Proteins/metabolism
14.
Nat Commun ; 15(1): 1021, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310114

ABSTRACT

The epidermal growth factor receptor (EGFR) plays important roles in multiple cellular events, including growth, differentiation, and motility. A major mechanism of downregulating EGFR function involves its endocytic transport to the lysosome. Sorting of proteins into intracellular pathways involves cargo adaptors recognizing sorting signals on cargo proteins. A dileucine-based sorting signal has been identified previously for the sorting of endosomal EGFR to the lysosome, but a cargo adaptor that recognizes this signal remains unknown. Here, we find that phosphoglycerate kinase 1 (PGK1) is recruited to endosomal membrane upon its phosphorylation, where it binds to the dileucine sorting signal in EGFR to promote the lysosomal transport of this receptor. We also elucidate two mechanisms that act in concert to promote PGK1 recruitment to endosomal membrane, a lipid-based mechanism that involves phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and a protein-based mechanism that involves hepatocyte growth factor receptor substrate (Hrs). These findings reveal an unexpected function for a metabolic enzyme and advance the mechanistic understanding of how EGFR is transported to the lysosome.


Subject(s)
ErbB Receptors , Phosphoglycerate Kinase , Phosphoglycerate Kinase/metabolism , ErbB Receptors/metabolism , Endosomes/metabolism , Proteins/metabolism , Lysosomes/metabolism , Protein Transport/physiology , Endosomal Sorting Complexes Required for Transport/metabolism
15.
bioRxiv ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38853868

ABSTRACT

Lipid transfer through membrane contact has been implicated to support vesicular transport, but a mechanistic understanding of this process remains to be achieved. Here, examining Coat Protein I (COPI) transport, we find that phosphatidylcholine (PC) with short acyl chains (sPC), which is needed to support COPI vesicle fission, is delivered through membrane contact from the endoplasmic reticulum (ER) to the Golgi complex at sites of COPI vesicle formation. Phosphatidylinositol transfer protein beta (PITPß) plays a central role in this delivery by not only catalyzing PC transfer, but also forming membrane contact. By combining cell-based studies with reconstitution approaches, we achieve spatial and temporal detail in explaining how sPC delivery occurs. Our findings advance the mechanistic understanding of how membrane contact is needed for vesicular transport in a model pathway and shed new insights into how PITPß acts.

16.
bioRxiv ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38562716

ABSTRACT

Cancer cell fate has been widely ascribed to mutational changes within protein-coding genes associated with tumor suppressors and oncogenes. In contrast, the mechanisms through which the biophysical properties of membrane lipids influence cancer cell survival, dedifferentiation and metastasis have received little scrutiny. Here, we report that cancer cells endowed with a high metastatic ability and cancer stem cell-like traits employ ether lipids to maintain low membrane tension and high membrane fluidity. Using genetic approaches and lipid reconstitution assays, we show that these ether lipid-regulated biophysical properties permit non-clathrin-mediated iron endocytosis via CD44, leading directly to significant increases in intracellular redox-active iron and enhanced ferroptosis susceptibility. Using a combination of in vitro three-dimensional microvascular network systems and in vivo animal models, we show that loss of ether lipids also strongly attenuates extravasation, metastatic burden and cancer stemness. These findings illuminate a mechanism whereby ether lipids in carcinoma cells serve as key regulators of malignant progression while conferring a unique vulnerability that can be exploited for therapeutic intervention.

17.
J Biol Chem ; 287(34): 28675-85, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22645133

ABSTRACT

Coat complexes sort protein cargoes into vesicular transport pathways. An emerging class of coat components has been the GTPase-activating proteins (GAPs) that act on the ADP-ribosylation factor (ARF) family of small GTPases. ACAP1 (ArfGAP with coiled-coil, ankyrin repeat, and PH domains protein 1) is an ARF6 GAP that also acts as a key component of a recently defined clathrin complex for endocytic recycling. Phosphorylation by Akt has been shown to enhance cargo binding by ACAP1 in explaining how integrin recycling is an example of regulated transport. We now shed further mechanistic insights into how this regulation is achieved at the level of cargo binding by ACAP1. We initially defined a critical sequence in the cytoplasmic domain of integrin ß1 recognized by ACAP1 and showed that this sequence acts as a recycling sorting signal. We then pursued a combination of structural, modeling, and functional studies, which suggest that phosphorylation of ACAP1 relieves a localized mechanism of autoinhibition in regulating cargo binding. Thus, we have elucidated a key regulatory juncture that controls integrin recycling and also advanced the understanding of how regulated cargo binding can lead to regulated transport.


Subject(s)
Clathrin/metabolism , GTPase-Activating Proteins/metabolism , Models, Biological , Protein Sorting Signals/physiology , Biological Transport, Active/physiology , Clathrin/genetics , GTPase-Activating Proteins/genetics , HeLa Cells , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Phosphorylation/physiology , Protein Binding/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
18.
Nat Cell Biol ; 8(12): 1376-82, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086176

ABSTRACT

Brefeldin-A ADP-ribosylated substrate (BARS) and dynamin function in membrane fission in distinct intracellular transport pathways, but whether their functions are mechanistically similar is unclear. Here, we show that ARFGAP1, a GTPase-activating protein (GAP) for ADP-ribosylation factor 1 (ARF1), couples to either BARS or endophilin B for vesicle formation by the coat protein I (COPI) complex - a finding that reveals an unanticipated mechanistic flexibility in mammalian COPI transport. Because dynamin is coupled to endophilin A in vesicle formation by the clathrin-coat complex, our finding also predicts that dynamin and ARF GAPs are likely to be functional counterparts in membrane fission among different transport pathways that connect intracellular membrane compartments.


Subject(s)
Intracellular Membranes/metabolism , Acyltransferases/metabolism , Animals , Coat Protein Complex I/metabolism , Coated Vesicles/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Golgi Apparatus/ultrastructure , Intracellular Membranes/ultrastructure , Mice , Transcription Factors/metabolism
19.
J Am Soc Nephrol ; 23(9): 1506-17, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22859853

ABSTRACT

The aquaporin 2 (AQP2) water channel, expressed in kidney collecting ducts, contributes critically to water homeostasis in mammals. Animals lacking or having significantly reduced levels of AQP2, however, have not only urinary concentrating abnormalities but also renal tubular defects that lead to neonatal mortality from renal failure. Here, we show that AQP2 is not only a water channel but also an integrin-binding membrane protein that promotes cell migration and epithelial morphogenesis. AQP2 expression modulates the trafficking and internalization of integrin ß1, facilitating its turnover at focal adhesions. In vitro, disturbing the interaction between AQP2 and integrin ß1 by mutating the RGD motif led to reduced endocytosis, retention of integrin ß1 at the cell surface, and defective cell migration and tubulogenesis. Similarly, in vivo, AQP2-null mice exhibited significant retention of integrin ß1 at the basolateral membrane and had tubular abnormalities. In summary, these data suggest that the water channel AQP2 interacts with integrins to promote renal epithelial cell migration, contributing to the structural and functional integrity of the mammalian kidney.


Subject(s)
Aquaporin 2/physiology , Cell Movement/physiology , Epithelial Cells/cytology , Kidney/cytology , Morphogenesis/physiology , Animals , Aquaporin 2/deficiency , Aquaporin 2/genetics , Cell Line , Cell Membrane Permeability/physiology , Dogs , Endocytosis/physiology , Epithelial Cells/physiology , In Vitro Techniques , Integrin beta1/physiology , Kidney/growth & development , Kidney/physiology , Mice , Mice, Knockout , Models, Animal , Mutation/genetics , Oligopeptides/genetics , Oligopeptides/physiology , Swine , Transfection
20.
Methods Mol Biol ; 2557: 519-528, 2023.
Article in English | MEDLINE | ID: mdl-36512234

ABSTRACT

The Coat Protein I (COPI) complex forms vesicles from Golgi membrane for retrograde transport among the Golgi stacks, and also from the Golgi to the endoplasmic reticulum (ER). We have been elucidating the mechanistic details of COPI vesicle formation through a reconstitution system that involves the incubation of Golgi membrane with purified components. This approach has enabled us recently to gain new insight into how certain lipids are critical for the fission stage of COPI vesicle formation. Lipid geometry has been proposed to act in the formation of transport carriers by promoting membrane curvature. However, evidence for this role has come from studies using simplified membranes, while confirmation in the more physiologic setting of native membranes has been challenging, as such membranes contain a complex composition of lipids and proteins. We have recently refined the COPI reconstitution system to overcome this experimental obstacle. This has led us to identify an unanticipated type of lipid geometry needed for COPI vesicle fission. This chapter describes the approach that we have developed to enable this discovery. The methodologies include: (i) preparation Golgi membrane from cells that are deficient in a particular lipid enzyme activity and (ii) functional rescue of this deficiency by introducing the product of the lipid enzyme, with experiments being performed at the in vitro level to gain mechanistic clarity and at the in vivo level to confirm physiologic relevance.


Subject(s)
COP-Coated Vesicles , Golgi Apparatus , COP-Coated Vesicles/metabolism , Golgi Apparatus/metabolism , Endoplasmic Reticulum/metabolism , Coat Protein Complex I/metabolism , Lipids
SELECTION OF CITATIONS
SEARCH DETAIL