Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Cell ; 185(13): 2292-2308.e20, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35750034

ABSTRACT

Lysosomes require an acidic lumen between pH 4.5 and 5.0 for effective digestion of macromolecules. This pH optimum is maintained by proton influx produced by the V-ATPase and efflux through an unidentified "H+ leak" pathway. Here we show that TMEM175, a genetic risk factor for Parkinson's disease (PD), mediates the lysosomal H+ leak by acting as a proton-activated, proton-selective channel on the lysosomal membrane (LyPAP). Acidification beyond the normal range potently activated LyPAP to terminate further acidification of lysosomes. An endogenous polyunsaturated fatty acid and synthetic agonists also activated TMEM175 to trigger lysosomal proton release. TMEM175 deficiency caused lysosomal over-acidification, impaired proteolytic activity, and facilitated α-synuclein aggregation in vivo. Mutational and pH normalization analyses indicated that the channel's H+ conductance is essential for normal lysosome function. Thus, modulation of LyPAP by cellular cues may dynamically tune the pH optima of endosomes and lysosomes to regulate lysosomal degradation and PD pathology.


Subject(s)
Parkinson Disease , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Lysosomes/metabolism , Parkinson Disease/metabolism , Potassium Channels/metabolism , Protons
2.
Physiol Rev ; 104(3): 1335-1385, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38451235

ABSTRACT

The endomembrane system consists of organellar membranes in the biosynthetic pathway [endoplasmic reticulum (ER), Golgi apparatus, and secretory vesicles] as well as those in the degradative pathway (early endosomes, macropinosomes, phagosomes, autophagosomes, late endosomes, and lysosomes). These endomembrane organelles/vesicles work together to synthesize, modify, package, transport, and degrade proteins, carbohydrates, and lipids, regulating the balance between cellular anabolism and catabolism. Large ion concentration gradients exist across endomembranes: Ca2+ gradients for most endomembrane organelles and H+ gradients for the acidic compartments. Ion (Na+, K+, H+, Ca2+, and Cl-) channels on the organellar membranes control ion flux in response to cellular cues, allowing rapid informational exchange between the cytosol and organelle lumen. Recent advances in organelle proteomics, organellar electrophysiology, and luminal and juxtaorganellar ion imaging have led to molecular identification and functional characterization of about two dozen endomembrane ion channels. For example, whereas IP3R1-3 channels mediate Ca2+ release from the ER in response to neurotransmitter and hormone stimulation, TRPML1-3 and TMEM175 channels mediate lysosomal Ca2+ and H+ release, respectively, in response to nutritional and trafficking cues. This review aims to summarize the current understanding of these endomembrane channels, with a focus on their subcellular localizations, ion permeation properties, gating mechanisms, cell biological functions, and disease relevance.


Subject(s)
Ion Channels , Humans , Animals , Ion Channels/metabolism , Intracellular Membranes/metabolism , Organelles/metabolism , Organelles/physiology
3.
Cell ; 161(6): 1306-19, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26027738

ABSTRACT

Vertebrate cells have evolved elaborate cell-autonomous defense programs to monitor subcellular compartments for infection and to evoke counter-responses. These programs are activated by pathogen-associated pattern molecules and by various strategies intracellular pathogens employ to alter cellular microenvironments. Here, we show that, when uropathogenic E. coli (UPEC) infect bladder epithelial cells (BECs), they are targeted by autophagy but avoid degradation because of their capacity to neutralize lysosomal pH. This change is detected by mucolipin TRP channel 3 (TRPML3), a transient receptor potential cation channel localized to lysosomes. TRPML3 activation then spontaneously initiates lysosome exocytosis, resulting in expulsion of exosome-encased bacteria. These studies reveal a cellular default system for lysosome homeostasis that has been co-opted by the autonomous defense program to clear recalcitrant pathogens.


Subject(s)
Escherichia coli Infections/immunology , Lysosomes/microbiology , TRPC Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/physiology , Animals , Autophagy , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Exocytosis , Lysosomes/enzymology , Lysosomes/metabolism , Mice , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology
4.
Cell ; 151(2): 372-83, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063126

ABSTRACT

Mammalian two-pore channel proteins (TPC1, TPC2; TPCN1, TPCN2) encode ion channels in intracellular endosomes and lysosomes and were proposed to mediate endolysosomal calcium release triggered by the second messenger, nicotinic acid adenine dinucleotide phosphate (NAADP). By directly recording TPCs in endolysosomes from wild-type and TPC double-knockout mice, here we show that, in contrast to previous conclusions, TPCs are in fact sodium-selective channels activated by PI(3,5)P(2) and are not activated by NAADP. Moreover, the primary endolysosomal ion is Na(+), not K(+), as had been previously assumed. These findings suggest that the organellar membrane potential may undergo large regulatory changes and may explain the specificity of PI(3,5)P(2) in regulating the fusogenic potential of intracellular organelles.


Subject(s)
Calcium Channels/metabolism , Lysosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , NADP/analogs & derivatives , NADP/metabolism , Sodium Channels/metabolism
5.
Cell ; 141(2): 331-43, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20403327

ABSTRACT

A plethora of growth factors regulate keratinocyte proliferation and differentiation that control hair morphogenesis and skin barrier formation. Wavy hair phenotypes in mice result from naturally occurring loss-of-function mutations in the genes for TGF-alpha and EGFR. Conversely, excessive activities of TGF-alpha/EGFR result in hairless phenotypes and skin cancers. Unexpectedly, we found that mice lacking the Trpv3 gene also exhibit wavy hair coat and curly whiskers. Here we show that keratinocyte TRPV3, a member of the transient receptor potential (TRP) family of Ca(2+)-permeant channels, forms a signaling complex with TGF-alpha/EGFR. Activation of EGFR leads to increased TRPV3 channel activity, which in turn stimulates TGF-alpha release. TRPV3 is also required for the formation of the skin barrier by regulating the activities of transglutaminases, a family of Ca(2+)-dependent crosslinking enzymes essential for keratinocyte cornification. Our results show that a TRP channel plays a role in regulating growth factor signaling by direct complex formation.


Subject(s)
ErbB Receptors/metabolism , Hair/growth & development , Signal Transduction , Skin/growth & development , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Hair/metabolism , Humans , Keratinocytes/metabolism , Mice , Mice, Knockout , Skin/metabolism , TRPV Cation Channels/genetics , Transforming Growth Factor alpha/metabolism
6.
PLoS Biol ; 19(7): e3001361, 2021 07.
Article in English | MEDLINE | ID: mdl-34297722

ABSTRACT

The lysosome is an essential organelle to recycle cellular materials and maintain nutrient homeostasis, but the mechanism to down-regulate its membrane proteins is poorly understood. In this study, we performed a cycloheximide (CHX) chase assay to measure the half-lives of approximately 30 human lysosomal membrane proteins (LMPs) and identified RNF152 and LAPTM4A as short-lived membrane proteins. The degradation of both proteins is ubiquitin dependent. RNF152 is a transmembrane E3 ligase that ubiquitinates itself, whereas LAPTM4A uses its carboxyl-terminal PY motifs to recruit NEDD4-1 for ubiquitination. After ubiquitination, they are internalized into the lysosome lumen by the endosomal sorting complexes required for transport (ESCRT) machinery for degradation. Strikingly, when ectopically expressed in budding yeast, human RNF152 is still degraded by the vacuole (yeast lysosome) in an ESCRT-dependent manner. Thus, our study uncovered a conserved mechanism to down-regulate lysosome membrane proteins.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Intracellular Membranes/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Humans , Proteolysis , Ubiquitin-Protein Ligases/metabolism
7.
Trends Biochem Sci ; 44(2): 110-124, 2019 02.
Article in English | MEDLINE | ID: mdl-30424907

ABSTRACT

Lysosomes, the degradation center of the cell, are filled with acidic hydrolases. Lysosomes generate nutrient-sensitive signals to regulate the import of H+, hydrolases, and endocytic and autophagic cargos, as well as the export of their degradation products (catabolites). In response to environmental and cellular signals, lysosomes change their positioning, number, morphology, size, composition, and activity within minutes to hours to meet the changing cellular needs. Ion channels in the lysosome are essential transducers that mediate signal-initiated Ca2+/Fe2+/Zn2+ release and H+/Na+/K+-dependent changes of membrane potential across the perimeter membrane. Dysregulation of lysosomal ion flux impairs lysosome movement, membrane trafficking, nutrient sensing, membrane repair, organelle membrane contact, and lysosome biogenesis and adaptation. Hence, activation and inhibition of lysosomal channels by synthetic modulators may tune lysosome function to maintain cellular health and promote cellular clearance in lysosome storage disorders.


Subject(s)
Ion Channels/metabolism , Lysosomes/metabolism , Signal Transduction , Animals , Cell Line , Humans , Ion Channels/chemistry , Lysosomes/chemistry , Models, Molecular , Particle Size
8.
J Cell Sci ; 134(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34254641

ABSTRACT

The transient receptor potential (TRP) channel superfamily consists of a large group of non-selective cation channels that serve as cellular sensors for a wide spectrum of physical and environmental stimuli. The 28 mammalian TRPs, categorized into six subfamilies, including TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPA (ankyrin), TRPML (mucolipin) and TRPP (polycystin), are widely expressed in different cells and tissues. TRPs exhibit a variety of unique features that not only distinguish them from other superfamilies of ion channels, but also confer diverse physiological functions. Located at the plasma membrane or in the membranes of intracellular organelles, TRPs are the cellular safeguards that sense various cell stresses and environmental stimuli and translate this information into responses at the organismal level. Loss- or gain-of-function mutations of TRPs cause inherited diseases and pathologies in different physiological systems, whereas up- or down-regulation of TRPs is associated with acquired human disorders. In this Cell Science at a Glance article and the accompanying poster, we briefly summarize the history of the discovery of TRPs, their unique features, recent advances in the understanding of TRP activation mechanisms, the structural basis of TRP Ca2+ selectivity and ligand binding, as well as potential roles in mammalian physiology and pathology.


Subject(s)
Transient Receptor Potential Channels , Animals , Humans , Ion Transport , Mammals/metabolism , Signal Transduction , TRPC Cation Channels , TRPP Cation Channels/metabolism , TRPV Cation Channels , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
9.
Nature ; 550(7676): 415-418, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29019981

ABSTRACT

Transient receptor potential mucolipin 1 (TRPML1) is a cation channel located within endosomal and lysosomal membranes. Ubiquitously expressed in mammalian cells, its loss-of-function mutations are the direct cause of type IV mucolipidosis, an autosomal recessive lysosomal storage disease. Here we present the single-particle electron cryo-microscopy structure of the mouse TRPML1 channel embedded in nanodiscs. Combined with mutagenesis analysis, the TRPML1 structure reveals that phosphatidylinositol-3,5-bisphosphate (PtdIns(3,5)P2) binds to the N terminus of the channel-distal from the pore-and the helix-turn-helix extension between segments S2 and S3 probably couples ligand binding to pore opening. The tightly packed selectivity filter contains multiple ion-binding sites, and the conserved acidic residues form the luminal Ca2+-blocking site that confers luminal pH and Ca2+ modulation on channel conductance. A luminal linker domain forms a fenestrated canopy atop the channel, providing several luminal ion passages to the pore and creating a negative electrostatic trap, with a preference for divalent cations, at the luminal entrance. The structure also reveals two equally distributed S4-S5 linker conformations in the closed channel, suggesting an S4-S5 linker-mediated PtdInsP2 gating mechanism among TRPML channels.


Subject(s)
Cryoelectron Microscopy , Endosomes/chemistry , Lysosomes/chemistry , Nanostructures/chemistry , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/ultrastructure , Animals , Binding Sites , Calcium , Hydrogen-Ion Concentration , Ion Transport , Ligands , Mice , Models, Molecular , Mutation , Phosphatidylinositol Phosphates/metabolism , Protein Conformation , Static Electricity , Transient Receptor Potential Channels/genetics
10.
Handb Exp Pharmacol ; 278: 93-108, 2023.
Article in English | MEDLINE | ID: mdl-36882602

ABSTRACT

Intracellular organelles exchange their luminal contents with each other via both vesicular and non-vesicular mechanisms. By forming membrane contact sites (MCSs) with ER and mitochondria, lysosomes mediate bidirectional transport of metabolites and ions between lysosomes and organelles that regulate lysosomal physiology, movement, membrane remodeling, and membrane repair. In this chapter, we will first summarize the current knowledge of lysosomal ion channels and then discuss the molecular and physiological mechanisms that regulate lysosome-organelle MCS formation and dynamics. We will also discuss the roles of lysosome-ER and lysosome-mitochondria MCSs in signal transduction, lipid transport, Ca 2+ transfer, membrane trafficking, and membrane repair, as well as their roles in lysosome-related pathologies.


Subject(s)
Ion Channels , Organelles , Humans , Organelles/metabolism , Lysosomes/metabolism , Mitochondria/metabolism , Biological Transport
11.
Proc Natl Acad Sci U S A ; 117(46): 29155-29165, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139539

ABSTRACT

LRRC8 family proteins on the plasma membrane play a critical role in cellular osmoregulation by forming volume-regulated anion channels (VRACs) necessary to prevent necrotic cell death. We demonstrate that intracellular LRRC8 proteins acting within lysosomes also play an essential role in cellular osmoregulation. LRRC8 proteins on lysosome membranes generate large lysosomal volume-regulated anion channel (Lyso-VRAC) currents in response to low cytoplasmic ionic strength conditions. When a double-leucine L706L707 motif at the C terminus of LRRC8A was mutated to alanines, normal plasma membrane VRAC currents were still observed, but Lyso-VRAC currents were absent. We used this targeting mutant, as well as pharmacological tools, to demonstrate that Lyso-VRAC currents are necessary for the formation of large lysosome-derived vacuoles, which store and then expel excess water to maintain cytosolic water homeostasis. Thus, Lyso-VRACs allow lysosomes of mammalian cells to act as the cell`s "bladder." When Lyso-VRAC current was selectively eliminated, the extent of necrotic cell death to sustained stress was greatly increased, not only in response to hypoosmotic stress, but also to hypoxic and hypothermic stresses. Thus Lyso-VRACs play an essential role in enabling cells to mount successful homeostatic responses to multiple stressors.


Subject(s)
Lysosomes/metabolism , Membrane Proteins/metabolism , Osmoregulation/physiology , Stress, Physiological/physiology , Animals , Anions , COS Cells , Cell Survival/physiology , Chlorocebus aethiops , Exocytosis , Gene Knockout Techniques , HEK293 Cells , Homeostasis , Humans , Membrane Proteins/genetics , Mice , Transcriptome , Vacuoles
12.
Proc Natl Acad Sci U S A ; 117(47): 29914-29924, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168737

ABSTRACT

Neuropeptides are important for regulating numerous neural functions and behaviors. Release of neuropeptides requires long-lasting, high levels of cytosolic Ca2+ However, the molecular regulation of neuropeptide release remains to be clarified. Recently, Stac3 was identified as a key regulator of L-type Ca2+ channels (CaChs) and excitation-contraction coupling in vertebrate skeletal muscles. There is a small family of stac genes in vertebrates with other members expressed by subsets of neurons in the central nervous system. The function of neural Stac proteins, however, is poorly understood. Drosophila melanogaster contain a single stac gene, Dstac, which is expressed by muscles and a subset of neurons, including neuropeptide-expressing motor neurons. Here, genetic manipulations, coupled with immunolabeling, Ca2+ imaging, electrophysiology, and behavioral analysis, revealed that Dstac regulates L-type CaChs (Dmca1D) in Drosophila motor neurons and this, in turn, controls the release of neuropeptides.


Subject(s)
Calcium Channels/metabolism , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Neuropeptides/metabolism , Animals , Animals, Genetically Modified , Behavior Observation Techniques , Behavior, Animal , Drosophila melanogaster , Female , Intravital Microscopy , Larva , Male , Models, Animal , Motor Neurons/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Neuromuscular Junction/cytology , Optical Imaging , Patch-Clamp Techniques , Presynaptic Terminals/metabolism
13.
PLoS Biol ; 17(5): e3000252, 2019 05.
Article in English | MEDLINE | ID: mdl-31112550

ABSTRACT

Rapamycin (Rap) and its derivatives, called rapalogs, are being explored in clinical trials targeting cancer and neurodegeneration. The underlying mechanisms of Rap actions, however, are not well understood. Mechanistic target of rapamycin (mTOR), a lysosome-localized protein kinase that acts as a critical regulator of cellular growth, is believed to mediate most Rap actions. Here, we identified mucolipin 1 (transient receptor potential channel mucolipin 1 [TRPML1], also known as MCOLN1), the principle Ca2+ release channel in the lysosome, as another direct target of Rap. Patch-clamping of isolated lysosomal membranes showed that micromolar concentrations of Rap and some rapalogs activated lysosomal TRPML1 directly and specifically. Pharmacological inhibition or genetic inactivation of mTOR failed to mimic the Rap effect. In vitro binding assays revealed that Rap bound directly to purified TRPML1 proteins with a micromolar affinity. In both healthy and disease human fibroblasts, Rap and rapalogs induced autophagic flux via nuclear translocation of transcription factor EB (TFEB). However, such effects were abolished in TRPML1-deficient cells or by TRPML1 inhibitors. Hence, Rap and rapalogs promote autophagy via a TRPML1-dependent mechanism. Given the demonstrated roles of TRPML1 and TFEB in cellular clearance, we propose that lysosomal TRPML1 may contribute a significant portion to the in vivo neuroprotective and anti-aging effects of Rap via an augmentation of autophagy and lysosomal biogenesis.


Subject(s)
Lysosomes/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transient Receptor Potential Channels/metabolism , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating/drug effects , Lysosomes/drug effects , Models, Biological , Protein Binding/drug effects , Sirolimus/analogs & derivatives , Sirolimus/chemistry
14.
Annu Rev Physiol ; 77: 57-80, 2015.
Article in English | MEDLINE | ID: mdl-25668017

ABSTRACT

Lysosomes are acidic compartments filled with more than 60 different types of hydrolases. They mediate the degradation of extracellular particles from endocytosis and of intracellular components from autophagy. The digested products are transported out of the lysosome via specific catabolite exporters or via vesicular membrane trafficking. Lysosomes also contain more than 50 membrane proteins and are equipped with the machinery to sense nutrient availability, which determines the distribution, number, size, and activity of lysosomes to control the specificity of cargo flux and timing (the initiation and termination) of degradation. Defects in degradation, export, or trafficking result in lysosomal dysfunction and lysosomal storage diseases (LSDs). Lysosomal channels and transporters mediate ion flux across perimeter membranes to regulate lysosomal ion homeostasis, membrane potential, catabolite export, membrane trafficking, and nutrient sensing. Dysregulation of lysosomal channels underlies the pathogenesis of many LSDs and possibly that of metabolic and common neurodegenerative diseases.


Subject(s)
Exocytosis/physiology , Ion Channels/physiology , Lysosomes/physiology , Animals , Homeostasis/physiology , Humans , Ions/metabolism , Lysosomal Storage Diseases/physiopathology , Membrane Potentials/physiology
15.
Gastroenterology ; 153(6): 1555-1567.e15, 2017 12.
Article in English | MEDLINE | ID: mdl-28859856

ABSTRACT

BACKGROUND & AIMS: The multiple endocrine neoplasia, type 1 (MEN1) locus encodes the nuclear protein and tumor suppressor menin. MEN1 mutations frequently cause neuroendocrine tumors such as gastrinomas, characterized by their predominant duodenal location and local metastasis at time of diagnosis. Diffuse gastrin cell hyperplasia precedes the appearance of MEN1 gastrinomas, which develop within submucosal Brunner's glands. We investigated how menin regulates expression of the gastrin gene and induces generation of submucosal gastrin-expressing cell hyperplasia. METHODS: Primary enteric glial cultures were generated from the VillinCre:Men1FL/FL:Sst-/- mice or C57BL/6 mice (controls), with or without inhibition of gastric acid by omeprazole. Primary enteric glial cells from C57BL/6 mice were incubated with gastrin and separated into nuclear and cytoplasmic fractions. Cells were incubated with forskolin and H89 to activate or inhibit protein kinase A (a family of enzymes whose activity depends on cellular levels of cyclic AMP). Gastrin was measured in blood, tissue, and cell cultures using an ELISA. Immunoprecipitation with menin or ubiquitin was used to demonstrate post-translational modification of menin. Primary glial cells were incubated with leptomycin b and MG132 to block nuclear export and proteasome activity, respectively. We obtained human duodenal, lymph node, and pancreatic gastrinoma samples, collected from patients who underwent surgery from 1996 through 2007 in the United States or the United Kingdom. RESULTS: Enteric glial cells that stained positive for glial fibrillary acidic protein (GFAP+) expressed gastrin de novo through a mechanism that required PKA. Gastrin-induced nuclear export of menin via cholecystokinin B receptor (CCKBR)-mediated activation of PKA. Once exported from the nucleus, menin was ubiquitinated and degraded by the proteasome. GFAP and other markers of enteric glial cells (eg, p75 and S100B), colocalized with gastrin in human duodenal gastrinomas. CONCLUSIONS: MEN1-associated gastrinomas, which develop in the submucosa, might arise from enteric glial cells through hormone-dependent PKA signaling. This pathway disrupts nuclear menin function, leading to hypergastrinemia and associated sequelae.


Subject(s)
Duodenum/metabolism , Gastrins/metabolism , Neuroglia/enzymology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Duodenal Neoplasms/enzymology , Duodenal Neoplasms/genetics , Duodenal Neoplasms/pathology , Duodenum/drug effects , Duodenum/pathology , Gastrinoma/enzymology , Gastrinoma/genetics , Gastrinoma/pathology , Gastrins/genetics , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Humans , Hyperplasia , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/drug effects , Proteasome Inhibitors/pharmacology , Proteolysis , Proto-Oncogene Proteins/genetics , Proton Pump Inhibitors/pharmacology , Receptor, Cholecystokinin B/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Time Factors , Ubiquitination
16.
Proc Natl Acad Sci U S A ; 112(11): E1373-81, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733853

ABSTRACT

Upon nutrient starvation, autophagy digests unwanted cellular components to generate catabolites that are required for housekeeping biosynthesis processes. A complete execution of autophagy demands an enhancement in lysosome function and biogenesis to match the increase in autophagosome formation. Here, we report that mucolipin-1 (also known as TRPML1 or ML1), a Ca(2+) channel in the lysosome that regulates many aspects of lysosomal trafficking, plays a central role in this quality-control process. By using Ca(2+) imaging and whole-lysosome patch clamping, lysosomal Ca(2+) release and ML1 currents were detected within hours of nutrient starvation and were potently up-regulated. In contrast, lysosomal Na(+)-selective currents were not up-regulated. Inhibition of mammalian target of rapamycin (mTOR) or activation of transcription factor EB (TFEB) mimicked a starvation effect in fed cells. The starvation effect also included an increase in lysosomal proteostasis and enhanced clearance of lysosomal storage, including cholesterol accumulation in Niemann-Pick disease type C (NPC) cells. However, this effect was not observed when ML1 was pharmacologically inhibited or genetically deleted. Furthermore, overexpression of ML1 mimicked the starvation effect. Hence, lysosomal adaptation to environmental cues such as nutrient levels requires mTOR/TFEB-dependent, lysosome-to-nucleus regulation of lysosomal ML1 channels and Ca(2+) signaling.


Subject(s)
Amino Acids/deficiency , Lysosomes/metabolism , Transient Receptor Potential Channels/metabolism , Up-Regulation , Amino Acids/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Cell Line , Cell Nucleus/metabolism , Cholesterol/metabolism , Gene Expression Regulation , Humans , Mutation/genetics , Niemann-Pick Diseases/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Biosynthesis , Protein Transport , Proteolysis , Sodium/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic
17.
Semin Cell Dev Biol ; 45: 24-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26519113

ABSTRACT

Resealing allows cells to mend damaged membranes rapidly when plasma membrane (PM) disruptions occur. Models of PM repair mechanisms include the "lipid-patch", "endocytic removal", and "macro-vesicle shedding" models, all of which postulate a dependence on local increases in intracellular Ca(2+) at injury sites. Multiple calcium sensors, including synaptotagmin (Syt) VII, dysferlin, and apoptosis-linked gene-2 (ALG-2), are involved in PM resealing, suggesting that Ca(2+) may regulate multiple steps of the repair process. Although earlier studies focused exclusively on external Ca(2+), recent studies suggest that Ca(2+) release from intracellular stores may also be important for PM resealing. Hence, depending on injury size and the type of injury, multiple sources of Ca(2+) may be recruited to trigger and orchestrate repair processes. In this review, we discuss the mechanisms by which the resealing process is promoted by vesicular Ca(2+) channels and Ca(2+) sensors that accumulate at damage sites.


Subject(s)
Calcium Signaling , Cell Membrane/physiology , Animals , Exocytosis , Humans , Lysosomes/physiology , Transient Receptor Potential Channels/metabolism , Wound Healing
18.
Plant Cell Physiol ; 58(7): 1185-1195, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28158631

ABSTRACT

Phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] is a low-abundance phospholipid known to be associated with a wide variety of physiological functions in plants. However, the localization and dynamics of PI(3,5)P2 in plant cells remain largely unknown, partially due to the lack of an effective fluorescent probe. Using Arabidopsis transgenic plant expressing the PI(3,5)P2-labeling fluorescent probe (tagRFP-ML1N*2) developed based on a tandem repeat of the cytosolic phosphoinositide-interacting domain (ML1N) of the mammalian lysosomal transient receptor potential cation channel, Mucolipin 1 (TRPML1), here we show that PI(3,5)P2 is predominantly localized on the limited membranes of the FAB1- and SNX1-positive late endosomes, but rarely localized on the membranes of plant vacuoles or trans-Golgi network/early endosomes of cortical cells of the root differentiation zone. The late endosomal localization of tagRFP-ML1N*2 is reduced or abolished by pharmacological inhibition or genetic knockdown of expression of genes encoding PI(3,5)P2-synthesizing enzymes, FAB1A/B, but markedly increased with FAB1A overexpression. Notably, reactive oxygen species (ROS) significantly increase late endosomal levels of PI(3,5)P2. Thus, tandem ML1N-based PI(3,5)P2 probes can reliably monitor intracellular dynamics of PI(3,5)P2 in Arabidopsis cells with less binding activity to other endomembrane organelles.


Subject(s)
Arabidopsis/metabolism , Fluorescent Dyes/metabolism , Phosphatidylinositol Phosphates/metabolism , Arabidopsis/cytology , Arabidopsis/genetics , Endosomes/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomes/metabolism , Microscopy, Confocal , Phosphatidylinositols/analysis , Plant Roots/cytology , Plant Roots/genetics , Plant Roots/metabolism , Plants, Genetically Modified , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins , Transport Vesicles/metabolism , Vacuoles/metabolism , trans-Golgi Network/metabolism , Red Fluorescent Protein
19.
Proc Natl Acad Sci U S A ; 110(52): 21165-70, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324172

ABSTRACT

Phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] is a low-abundance phosphoinositide presumed to be localized to endosomes and lysosomes, where it recruits cytoplasmic peripheral proteins and regulates endolysosome-localized membrane channel activity. Cells lacking PI(3,5)P2 exhibit lysosomal trafficking defects, and human mutations in the PI(3,5)P2-metabolizing enzymes cause lysosome-related diseases. The spatial and temporal dynamics of PI(3,5)P2, however, remain unclear due to the lack of a reliable detection method. Of the seven known phosphoinositides, only PI(3,5)P2 binds, in the low nanomolar range, to a cytoplasmic phosphoinositide-interacting domain (ML1N) to activate late endosome and lysosome (LEL)-localized transient receptor potential Mucolipin 1 (TRPML1) channels. Here, we report the generation and characterization of a PI(3,5)P2-specific probe, generated by the fusion of fluorescence tags to the tandem repeats of ML1N. The probe was mainly localized to the membranes of Lamp1-positive compartments, and the localization pattern was dynamically altered by either mutations in the probe, or by genetically or pharmacologically manipulating the cellular levels of PI(3,5)P2. Through the use of time-lapse live-cell imaging, we found that the localization of the PI(3,5)P2 probe was regulated by serum withdrawal/addition, undergoing rapid changes immediately before membrane fusion of two LELs. Our development of a PI(3,5)P2-specific probe may facilitate studies of both intracellular signal transduction and membrane trafficking in the endosomes and lysosomes.


Subject(s)
Fluorescent Dyes/metabolism , Molecular Imaging/methods , Phosphatidylinositol Phosphates/metabolism , Transient Receptor Potential Channels/metabolism , Image Processing, Computer-Assisted , Microscopy, Confocal , Protein Binding , Transient Receptor Potential Channels/genetics , Transport Vesicles/metabolism
20.
J Neurosci ; 34(34): 11485-503, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143627

ABSTRACT

Antiretroviral therapy extends the lifespan of human immunodeficiency virus (HIV)-infected patients, but many survivors develop premature impairments in cognition. These residual cognitive impairments may involve aberrant deposition of amyloid ß-peptides (Aß). By unknown mechanisms, Aß accumulates in the lysosomal and autophagic compartments of neurons in the HIV-infected brain. Here we identify the molecular events evoked by the HIV coat protein gp120 that facilitate the intraneuronal accumulation of Aß. We created a triple transgenic gp120/APP/PS1 mouse that recapitulates intraneuronal deposition of Aß in a manner reminiscent of the HIV-infected brain. In cultured neurons, we found that the HIV coat protein gp120 increased the transcriptional expression of BACE1 through repression of PPARγ, and increased APP expression by promoting interaction of the translation-activating RBP heterogeneous nuclear ribonucleoprotein C with APP mRNA. APP and BACE1 were colocalized into stabilized membrane microdomains, where the ß-cleavage of APP and Aß formation were enhanced. Aß-peptides became localized to lysosomes that were engorged with sphingomyelin and calcium. Stimulating calcium efflux from lysosomes with a TRPM1 agonist promoted calcium efflux, luminal acidification, and cleared both sphingomyelin and Aß from lysosomes. These findings suggest that therapeutics targeted to reduce lysosomal pH in neurodegenerative conditions may protect neurons by facilitating the clearance of accumulated sphingolipids and Aß-peptides.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/pathology , HIV Infections/pathology , Neurons/metabolism , Transient Receptor Potential Channels/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/virology , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/pharmacology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Presenilin-1/genetics , Rats , Signal Transduction/drug effects , Transient Receptor Potential Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL