Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Cell Mol Life Sci ; 81(1): 29, 2024 Jan 12.
Article En | MEDLINE | ID: mdl-38212474

Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 h of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6, and TGFß3, all of which appear to be upregulated by increased intracellular calcium. We further demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis through a process involving inhibition of CDK4/6 and cell cycle progression. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.


Calcium , Milk , Female , Animals , Milk/metabolism , Calcium/metabolism , Cell Death , Lactation , Lysosomes/metabolism , Mammary Glands, Animal/metabolism , STAT3 Transcription Factor/metabolism
2.
Dev Cell ; 58(19): 1847-1863.e12, 2023 10 09.
Article En | MEDLINE | ID: mdl-37751746

An actin-spectrin lattice, the membrane periodic skeleton (MPS), protects axons from breakage. MPS integrity relies on spectrin delivery via slow axonal transport, a process that remains poorly understood. We designed a probe to visualize endogenous spectrin dynamics at single-axon resolution in vivo. Surprisingly, spectrin transport is bimodal, comprising fast runs and movements that are 100-fold slower than previously reported. Modeling and genetic analysis suggest that the two rates are independent, yet both require kinesin-1 and the coiled-coil proteins UNC-76/FEZ1 and UNC-69/SCOC, which we identify as spectrin-kinesin adaptors. Knockdown of either protein led to disrupted spectrin motility and reduced distal MPS, and UNC-76 overexpression instructed excessive transport of spectrin. Artificially linking spectrin to kinesin-1 drove robust motility but inefficient MPS assembly, whereas impairing MPS assembly led to excessive spectrin transport, suggesting a balance between transport and assembly. These results provide insight into slow axonal transport and MPS integrity.


Caenorhabditis elegans Proteins , Caenorhabditis elegans , Spectrin , Animals , Axonal Transport , Axons/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Kinesins/metabolism , Spectrin/metabolism
3.
Res Sq ; 2023 Jun 16.
Article En | MEDLINE | ID: mdl-37398309

Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 hours of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6 and TGFß3, all of which appear to be upregulated by increased intracellular calcium. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis. This is the result of increased TGFß signaling and inhibition of cell cycle progression. Finally, we demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3, a process which also appears to be mediated by TGFß signaling. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.

4.
Proc Natl Acad Sci U S A ; 120(31): e2303789120, 2023 08.
Article En | MEDLINE | ID: mdl-37487100

Cells maintain optimal levels of lysosome degradative activity to protect against pathogens, clear waste, and generate nutrients. Here, we show that LRRK2, a protein that is tightly linked to Parkinson's disease, negatively regulates lysosome degradative activity in macrophages and microglia via a transcriptional mechanism. Depletion of LRRK2 and inhibition of LRRK2 kinase activity enhanced lysosomal proteolytic activity and increased the expression of multiple lysosomal hydrolases. Conversely, the kinase hyperactive LRRK2 G2019S Parkinson's disease mutant suppressed lysosomal degradative activity and gene expression. We identified MiT-TFE transcription factors (TFE3, TFEB, and MITF) as mediators of LRRK2-dependent control of lysosomal gene expression. LRRK2 negatively regulated the abundance and nuclear localization of these transcription factors and their depletion prevented LRRK2-dependent changes in lysosome protein levels. These observations define a role for LRRK2 in controlling lysosome degradative activity and support a model wherein LRRK2 hyperactivity may increase Parkinson's disease risk by suppressing lysosome degradative activity.


Parkinson Disease , Transcription Factors , Humans , Transcription Factors/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Microglia/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , Macrophages/metabolism
5.
Traffic ; 24(1): 4-19, 2023 01.
Article En | MEDLINE | ID: mdl-36398980

The trans-Golgi Network (TGN) sorts molecular "addresses" and sends newly synthesized proteins to their destination via vesicular transport carriers. Despite the functional significance of packaging processes at the TGN, the sorting of soluble proteins remains poorly understood. Recent research has shown that the Golgi resident protein Cab45 is a significant regulator of secretory cargo sorting at the TGN. Cab45 oligomerizes upon transient Ca2+ influx, recruits soluble cargo molecules (clients), and packs them in sphingomyelin-rich transport carriers. However, the identity of client molecules packed into Cab45 vesicles is scarce. Therefore, we used a precise and highly efficient secretome analysis technology called hiSPECs. Intriguingly, we observed that Cab45 deficient cells manifest hypersecretion of lysosomal hydrolases. Specifically, Cab45 deficient cells secrete the unprocessed precursors of prosaposin (PSAP) and progranulin (PGRN). In addition, lysosomes in these cells show an aberrant perinuclear accumulation suggesting a new role of Cab45 in lysosomal positioning. This work uncovers a yet unknown function of Cab45 in regulating lysosomal function.


Proteins , Saposins , Humans , Biological Transport , Lysosomes/metabolism , Progranulins/metabolism , Protein Transport/physiology , Proteins/metabolism , Saposins/genetics , Saposins/metabolism , trans-Golgi Network/metabolism
6.
bioRxiv ; 2023 Dec 17.
Article En | MEDLINE | ID: mdl-38168426

Lysosomes play a pivotal role in coordinating macromolecule degradation and regulating cell growth and metabolism. Despite substantial progress in identifying lysosomal signaling proteins, understanding the pathways that synchronize lysosome functions with changing cellular demands remains incomplete. This study uncovers a role for TANK-binding kinase 1 (TBK1), well known for its role in innate immunity and organelle quality control, in modulating lysosomal responsiveness to nutrients. Specifically, we identify a pool of TBK1 that is recruited to lysosomes in response to elevated amino acid levels. At lysosomes, this TBK1 phosphorylates Rab7 on serine 72. This is critical for alleviating Rab7-mediated inhibition of amino acid-dependent mTORC1 activation. Furthermore, a TBK1 mutant (E696K) associated with amyotrophic lateral sclerosis and frontotemporal dementia constitutively accumulates at lysosomes, resulting in elevated Rab7 phosphorylation and increased mTORC1 activation. This data establishes the lysosome as a site of amino acid regulated TBK1 signaling that is crucial for efficient mTORC1 activation. This lysosomal pool of TBK1 has broader implications for lysosome homeostasis, and its dysregulation could contribute to the pathogenesis of ALS-FTD.

7.
J Cell Biol ; 221(7)2022 07 04.
Article En | MEDLINE | ID: mdl-35657605

Mutations in VPS13C cause early-onset, autosomal recessive Parkinson's disease (PD). We have established that VPS13C encodes a lipid transfer protein localized to contact sites between the ER and late endosomes/lysosomes. In the current study, we demonstrate that depleting VPS13C in HeLa cells causes an accumulation of lysosomes with an altered lipid profile, including an accumulation of di-22:6-BMP, a biomarker of the PD-associated leucine-rich repeat kinase 2 (LRRK2) G2019S mutation. In addition, the DNA-sensing cGAS-STING pathway, which was recently implicated in PD pathogenesis, is activated in these cells. This activation results from a combination of elevated mitochondrial DNA in the cytosol and a defect in the degradation of activated STING, a lysosome-dependent process. These results suggest a link between ER-lysosome lipid transfer and innate immune activation in a model human cell line and place VPS13C in pathways relevant to PD pathogenesis.


DNA, Mitochondrial , Endoplasmic Reticulum , Membrane Proteins , Parkinson Disease , Proteins , DNA, Mitochondrial/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lipids , Lysosomes/metabolism , Membrane Proteins/metabolism , Mutation , Parkinson Disease/metabolism , Proteins/metabolism
8.
J Cell Biol ; 221(2)2022 02 07.
Article En | MEDLINE | ID: mdl-34919127

Progranulin is a lysosomal protein whose haploinsufficiency causes frontotemporal dementia, while homozygous loss of progranulin causes neuronal ceroid lipofuscinosis, a lysosomal storage disease. The sensitivity of cells to progranulin deficiency raises important questions about how cells coordinate intracellular trafficking of progranulin to ensure its efficient delivery to lysosomes. In this study, we discover that progranulin interactions with prosaposin, another lysosomal protein, first occur within the lumen of the endoplasmic reticulum (ER) and are required for the efficient ER exit of progranulin. Mechanistically, we identify an interaction between prosaposin and Surf4, a receptor that promotes loading of lumenal cargos into COPII-coated vesicles, and establish that Surf4 is critical for the efficient export of progranulin and prosaposin from the ER. Collectively, this work demonstrates that a network of interactions occurring early in the secretory pathway promote the ER exit and subsequent lysosomal delivery of newly translated progranulin and prosaposin.


Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Progranulins/metabolism , Saposins/metabolism , Amino Acid Motifs , Animals , HeLa Cells , Humans , Mice , Protein Binding , Saposins/chemistry
9.
Nat Commun ; 12(1): 4245, 2021 07 12.
Article En | MEDLINE | ID: mdl-34253722

Tuberous Sclerosis Complex (TSC) is caused by TSC1 or TSC2 mutations, resulting in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). Transcription factor EB (TFEB), a master regulator of lysosome biogenesis, is negatively regulated by mTORC1 through a RAG GTPase-dependent phosphorylation. Here we show that lysosomal biogenesis is increased in TSC-associated renal tumors, pulmonary lymphangioleiomyomatosis, kidneys from Tsc2+/- mice, and TSC1/2-deficient cells via a TFEB-dependent mechanism. Interestingly, in TSC1/2-deficient cells, TFEB is hypo-phosphorylated at mTORC1-dependent sites, indicating that mTORC1 is unable to phosphorylate TFEB in the absence of the TSC1/2 complex. Importantly, overexpression of folliculin (FLCN), a GTPase activating protein for RAGC, increases TFEB phosphorylation at the mTORC1 sites in TSC2-deficient cells. Overexpression of constitutively active RAGC is sufficient to relocalize TFEB to the cytoplasm. These findings establish the TSC proteins as critical regulators of lysosomal biogenesis via TFEB and RAGC and identify TFEB as a driver of the proliferation of TSC2-deficient cells.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Organelle Biogenesis , Tuberous Sclerosis Complex 2 Protein/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinoma, Renal Cell/pathology , Cell Nucleus/metabolism , Cell Proliferation , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Kidney Neoplasms/pathology , Lysosomes/ultrastructure , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , Tuberous Sclerosis Complex 2 Protein/deficiency , Tumor Suppressor Proteins/metabolism
10.
PLoS Genet ; 17(4): e1009406, 2021 04.
Article En | MEDLINE | ID: mdl-33830999

Phospholipase D3 (PLD3) is a protein of unclear function that structurally resembles other members of the phospholipase D superfamily. A coding variant in this gene confers increased risk for the development of Alzheimer's disease (AD), although the magnitude of this effect has been controversial. Because of the potential significance of this obscure protein, we undertook a study to observe its distribution in normal human brain and AD-affected brain, determine whether PLD3 is relevant to memory and cognition in sporadic AD, and to evaluate its molecular function. In human neuropathological samples, PLD3 was primarily found within neurons and colocalized with lysosome markers (LAMP2, progranulin, and cathepsins D and B). This colocalization was also present in AD brain with prominent enrichment on lysosomal accumulations within dystrophic neurites surrounding ß-amyloid plaques. This pattern of protein distribution was conserved in mouse brain in wild type and the 5xFAD mouse model of cerebral ß-amyloidosis. We discovered PLD3 has phospholipase D activity in lysosomes. A coding variant in PLD3 reported to confer AD risk significantly reduced enzymatic activity compared to wild-type PLD3. PLD3 mRNA levels in the human pre-frontal cortex inversely correlated with ß-amyloid pathology severity and rate of cognitive decline in 531 participants enrolled in the Religious Orders Study and Rush Memory and Aging Project. PLD3 levels across genetically diverse BXD mouse strains and strains crossed with 5xFAD mice correlated strongly with learning and memory performance in a fear conditioning task. In summary, this study identified a new functional mammalian phospholipase D isoform which is lysosomal and closely associated with both ß-amyloid pathology and cognition.


Alzheimer Disease/genetics , Cognitive Dysfunction/genetics , Genetic Predisposition to Disease , Phospholipase D/genetics , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Animals , Autopsy , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/pathology , Disease Models, Animal , HeLa Cells , Humans , Lysosomes/enzymology , Lysosomes/pathology , Mice , Neurons/enzymology , Neurons/pathology , Plaque, Amyloid/enzymology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology
11.
Mol Biol Cell ; 32(11): 1094-1103, 2021 05 15.
Article En | MEDLINE | ID: mdl-33788575

The dependence of neurons on microtubule-based motors for the movement of lysosomes over long distances raises questions about adaptations that allow neurons to meet these demands. Recently, JIP3/MAPK8IP3, a neuronally enriched putative adaptor between lysosomes and motors, was identified as a critical regulator of axonal lysosome abundance. In this study, we establish a human induced pluripotent stem cell (iPSC)-derived neuron model for the investigation of axonal lysosome transport and maturation and show that loss of JIP3 results in the accumulation of axonal lysosomes and the Alzheimer's disease-related amyloid precursor protein (APP)-derived Aß42 peptide. We furthermore reveal an overlapping role of the homologous JIP4 gene in lysosome axonal transport. These results establish a cellular model for investigating the relationship between lysosome axonal transport and amyloidogenic APP processing and more broadly demonstrate the utility of human iPSC-derived neurons for the investigation of neuronal cell biology and pathology.


Adaptor Proteins, Signal Transducing/physiology , Axonal Transport/physiology , Nerve Tissue Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Axons/metabolism , Axons/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Lysosomes/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/physiology , Protein Transport/physiology
12.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article En | MEDLINE | ID: mdl-33597295

PQLC2, a lysosomal cationic amino acid transporter, also serves as a sensor that responds to scarcity of its substrates by recruiting a protein complex composed of C9orf72, SMCR8, and WDR41 to the surface of lysosomes. This protein complex controls multiple aspects of lysosome function. Although it is known that this response to changes in cationic amino acid availability depends on an interaction between PQLC2 and WDR41, the underlying mechanism for the regulated interaction is not known. In this study, we present evidence that the WDR41-PQLC2 interaction is mediated by a short peptide motif in a flexible loop that extends from the WDR41 ß-propeller and inserts into a cavity presented by the inward-facing conformation of PQLC2. The data support a transceptor model wherein conformational changes in PQLC2 related to substrate transport regulate the availability of the WDR41-binding site on PQLC2 and mediate recruitment of the WDR41-SMCR8-C9orf72 complex to the surface of lysosomes.


Amino Acid Transport Systems, Basic/metabolism , Amino Acids/metabolism , Lysosomes/metabolism , Amino Acid Motifs , Amino Acid Transport Systems, Basic/chemistry , Amino Acid Transport Systems, Basic/genetics , Amino Acids/chemistry , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Binding Sites , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , HeLa Cells , Humans , Multiprotein Complexes/metabolism , Mutagenesis , Protein Conformation , Protein Interaction Domains and Motifs
13.
F1000Res ; 92020.
Article En | MEDLINE | ID: mdl-32518628

A complex molecular machinery converges on the surface of lysosomes to ensure that the growth-promoting signaling mediated by mechanistic target of rapamycin complex 1 (mTORC1) is tightly controlled by the availability of nutrients and growth factors. The final step in this activation process is dependent on Rheb, a small GTPase that binds to mTOR and allosterically activates its kinase activity. Here we review the mechanisms that determine the subcellular localization of Rheb (and the closely related RhebL1 protein) as well as the significance of these mechanisms for controlling mTORC1 activation. In particular, we explore how the relatively weak membrane interactions conferred by C-terminal farnesylation are critical for the ability of Rheb to activate mTORC1. In addition to supporting transient membrane interactions, Rheb C-terminal farnesylation also supports an interaction between Rheb and the δ subunit of phosphodiesterase 6 (PDEδ). This interaction provides a potential mechanism for targeting Rheb to membranes that contain Arl2, a small GTPase that triggers the release of prenylated proteins from PDEδ. The minimal membrane targeting conferred by C-terminal farnesylation of Rheb and RhebL1 distinguishes them from other members of the Ras superfamily that possess additional membrane interaction motifs that work with farnesylation for enrichment on the specific subcellular membranes where they engage key effectors. Finally, we highlight diversity in Rheb membrane targeting mechanisms as well as the potential for alternative mTORC1 activation mechanisms across species.


Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Signal Transduction , Humans , Intercellular Signaling Peptides and Proteins , ras Proteins/metabolism
14.
J Cell Biol ; 219(1)2020 01 06.
Article En | MEDLINE | ID: mdl-31851326

The C9orf72 protein is required for normal lysosome function. In support of such functions, C9orf72 forms a heterotrimeric complex with SMCR8 and WDR41 that is recruited to lysosomes when amino acids are scarce. These properties raise questions about the identity of the lysosomal binding partner of the C9orf72 complex and the amino acid-sensing mechanism that regulates C9orf72 complex abundance on lysosomes. We now demonstrate that an interaction with the lysosomal cationic amino acid transporter PQLC2 mediates C9orf72 complex recruitment to lysosomes. This is achieved through an interaction between PQLC2 and WDR41. The interaction between PQLC2 and the C9orf72 complex is negatively regulated by arginine, lysine, and histidine, the amino acids that PQLC2 transports across the membrane of lysosomes. These results define a new role for PQLC2 in the regulated recruitment of the C9orf72 complex to lysosomes and reveal a novel mechanism that allows cells to sense and respond to changes in the availability of cationic amino acids within lysosomes.


Amino Acid Transport Systems, Basic/metabolism , Amino Acids/deficiency , C9orf72 Protein/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Amino Acid Transport Systems, Basic/genetics , Autophagy , C9orf72 Protein/genetics , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Protein Interaction Domains and Motifs
15.
Am J Hum Genet ; 105(5): 1023-1029, 2019 11 07.
Article En | MEDLINE | ID: mdl-31630788

We describe unrelated individuals with ichthyosis, failure to thrive, thrombocytopenia, photophobia, and progressive hearing loss. Each have bi-allelic mutations in AP1B1, the gene encoding the ß subunit of heterotetrameric adaptor protein 1 (AP-1) complexes, which mediate endomembrane polarization, sorting, and transport. In affected keratinocytes the AP-1 ß subunit is lost, and the γ subunit is greatly reduced, demonstrating destabilization of the AP-1 complex. Affected cells and tissue contain an abundance of abnormal vesicles and show hyperproliferation, abnormal epidermal differentiation, and derangement of intercellular junction proteins. Transduction of affected cells with wild-type AP1B1 rescues the vesicular phenotype, conclusively establishing that loss of AP1B1 function causes this disorder.


Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits/genetics , Deafness/genetics , Genes, Recessive/genetics , Ichthyosis/genetics , Mutation/genetics , Photophobia/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Female , Hearing Loss/genetics , Humans , Male , Phenotype , Protein Subunits/genetics , Protein Transport/genetics , Thrombocytopenia/genetics
16.
Life Sci Alliance ; 2(5)2019 10.
Article En | MEDLINE | ID: mdl-31527135

TDP-43 is an RNA-binding protein that forms cytoplasmic aggregates in multiple neurodegenerative diseases. Although the loss of normal TDP-43 functions likely contributes to disease pathogenesis, the cell biological consequences of human TDP-43 depletion are not well understood. We, therefore, generated human TDP-43 knockout (KO) cells and subjected them to parallel cell biological and transcriptomic analyses. These efforts yielded three important discoveries. First, complete loss of TDP-43 resulted in widespread morphological defects related to multiple organelles, including Golgi, endosomes, lysosomes, mitochondria, and the nuclear envelope. Second, we identified a new role for TDP-43 in controlling mRNA splicing of Nup188 (nuclear pore protein). Third, analysis of multiple amyotrophic lateral sclerosis causing TDP-43 mutations revealed a broad ability to support splicing of TDP-43 target genes. However, as some TDP-43 disease-causing mutants failed to fully support the regulation of specific target transcripts, our results raise the possibility of mutation-specific loss-of-function contributions to disease pathology.


Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling/methods , Nuclear Pore Complex Proteins/genetics , Organelles/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Genetic Pleiotropy , HeLa Cells , Homeostasis , Humans , Mutation , RNA Transport
17.
Mol Biol Cell ; 30(22): 2750-2760, 2019 10 15.
Article En | MEDLINE | ID: mdl-31532697

Stable localization of the Rheb GTPase to lysosomes is thought to be required for activation of mTOR complex 1 (mTORC1) signaling. However, the lysosome targeting mechanisms for Rheb remain unclear. We therefore investigated the relationship between Rheb subcellular localization and mTORC1 activation. Surprisingly, we found that Rheb was undetectable at lysosomes. Nonetheless, functional assays in knockout human cells revealed that farnesylation of the C-terminal CaaX motif on Rheb was essential for Rheb-dependent mTORC1 activation. Although farnesylated Rheb exhibited partial endoplasmic reticulum (ER) localization, constitutively targeting Rheb to ER membranes did not support mTORC1 activation. Further systematic analysis of Rheb lipidation revealed that weak, nonselective, membrane interactions support Rheb-dependent mTORC1 activation without the need for a specific lysosome targeting motif. Collectively, these results argue against stable interactions of Rheb with lysosomes and instead that transient membrane interactions optimally allow Rheb to activate mTORC1 signaling.


Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Amino Acids , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Lysosomes/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Monomeric GTP-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Neuropeptides/metabolism , Prenylation , Ras Homolog Enriched in Brain Protein/physiology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
18.
Cell Stem Cell ; 24(2): 199-200, 2019 02 07.
Article En | MEDLINE | ID: mdl-30735644

In this issue of Cell Stem Cell, Villegas et al. (2019) used a genome-wide CRISPR screen to identify ESC pluripotency regulators, which generated insights into how signaling from lysosome enables cells to transcriptionally regulate their metabolism. Further investigation of human genetics identified a human developmental disease arising from a defect in this process.


Embryonic Stem Cells , Pluripotent Stem Cells , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation , Genome , Humans , Lysosomes
19.
Neurosci Lett ; 697: 1-9, 2019 04 01.
Article En | MEDLINE | ID: mdl-29626653

Lysosomes support diverse cellular functions by acting as sites of macromolecule degradation and nutrient recycling. The degradative abilities of lysosomes are conferred by a lumen that is characterized by an acidic pH and which contains numerous hydrolases that support the breakdown of major cellular macromolecules to yield cellular building blocks (amino acids, nucleic acids, sugars, lipids and metals) that are transported into the cytoplasm for their re-use. In addition to these important hydrolytic and recycling functions, lysosomes also serve as a signaling platform that integrates nutrient and metabolic cues to control signaling via the mTORC1 pathway. Due to their extreme longevity, polarity, demands of neurotransmission and metabolic activity, neurons are particularly sensitive to perturbations in lysosome function. The dependence of neurons on optimal lysosome function is highlighted by insights from human genetics that link lysosome dysfunction to a wide range of both rare and common neurological diseases. How then is lysosome function adapted to the unique demands of neurons? This review will focus on the roles played by lysosomes in distinct neuronal sub-compartments, the regulation of neuronal lysosome sub-cellular localization and the implications of such neuronal lysosome regulation for both physiology and disease.


Lysosomes/metabolism , Neurons/metabolism , Animals , Autophagy/physiology , Humans , Neurons/cytology
20.
Mol Biol Cell ; 29(18): 2213-2227, 2018 09 01.
Article En | MEDLINE | ID: mdl-29995611

C9orf72 mutations are a major cause of amyotrophic lateral sclerosis and frontotemporal dementia. The C9orf72 protein undergoes regulated recruitment to lysosomes and has been broadly implicated in control of lysosome homeostasis. However, although evidence strongly supports an important function for C9orf72 at lysosomes, little is known about the lysosome recruitment mechanism. In this study, we identify an essential role for WDR41, a prominent C9orf72 interacting protein, in C9orf72 lysosome recruitment. Analysis of human WDR41 knockout cells revealed that WDR41 is required for localization of the protein complex containing C9orf72 and SMCR8 to lysosomes. Such lysosome localization increases in response to amino acid starvation but is not dependent on either mTORC1 inhibition or autophagy induction. Furthermore, WDR41 itself exhibits a parallel pattern of regulated association with lysosomes. This WDR41-dependent recruitment of C9orf72 to lysosomes is critical for the ability of lysosomes to support mTORC1 signaling as constitutive targeting of C9orf72 to lysosomes relieves the requirement for WDR41 in mTORC1 activation. Collectively, this study reveals an essential role for WDR41 in supporting the regulated binding of C9orf72 to lysosomes and solidifies the requirement for a larger C9orf72 containing protein complex in coordinating lysosomal responses to changes in amino acid availability.


Amino Acids/metabolism , Carrier Proteins/metabolism , Lysosomes/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Autophagy , Autophagy-Related Proteins , C9orf72 Protein/metabolism , Carrier Proteins/genetics , Frontotemporal Dementia/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mutation , Signal Transduction
...