Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.446
Filter
Add more filters

Publication year range
1.
Cell ; 169(4): 766-766.e1, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475901

ABSTRACT

Macropinocytosis is the bulk ingestion of extracellular fluids via large endocytic vacuoles. This SnapShot provides an overview of physiological macropinocytosis in immune surveillance and its pathogenic contribution during infection and cancer proliferation.


Subject(s)
Pinocytosis , Animals , Endocytosis , Humans , Immunologic Surveillance , Infections/immunology , Vacuoles
2.
Mol Cell ; 84(11): 2104-2118.e6, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38761795

ABSTRACT

Circular RNAs (circRNAs) are stable RNAs present in cell-free RNA, which may comprise cellular debris and pathogen genomes. Here, we investigate the phenomenon and mechanism of cellular uptake and intracellular fate of exogenous circRNAs. Human myeloid cells and B cells selectively internalize extracellular circRNAs. Macrophage uptake of circRNA is rapid, energy dependent, and saturable. CircRNA uptake can lead to translation of encoded sequences and antigen presentation. The route of internalization influences immune activation after circRNA uptake, with distinct gene expression programs depending on the route of RNA delivery. Genome-scale CRISPR screens and chemical inhibitor studies nominate macrophage scavenger receptor MSR1, Toll-like receptors, and mTOR signaling as key regulators of receptor-mediated phagocytosis of circRNAs, a dominant pathway to internalize circRNAs in parallel to macropinocytosis. These results suggest that cell-free circRNA serves as an "eat me" signal and danger-associated molecular pattern, indicating orderly pathways of recognition and disposal.


Subject(s)
Macrophages , Phagocytosis , RNA, Circular , Signal Transduction , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Macrophages/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Animals , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class A/genetics , Antigen Presentation , Pinocytosis , Mice
3.
Cell ; 162(2): 259-270, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26144316

ABSTRACT

Despite being surrounded by diverse nutrients, mammalian cells preferentially metabolize glucose and free amino acids. Recently, Ras-induced macropinocytosis of extracellular proteins was shown to reduce a transformed cell's dependence on extracellular glutamine. Here, we demonstrate that protein macropinocytosis can also serve as an essential amino acid source. Lysosomal degradation of extracellular proteins can sustain cell survival and induce activation of mTORC1 but fails to elicit significant cell accumulation. Unlike its growth-promoting activity under amino-acid-replete conditions, we discovered that mTORC1 activation suppresses proliferation when cells rely on extracellular proteins as an amino acid source. Inhibiting mTORC1 results in increased catabolism of endocytosed proteins and enhances cell proliferation during nutrient-depleted conditions in vitro and within vascularly compromised tumors in vivo. Thus, by preventing nutritional consumption of extracellular proteins, mTORC1 couples growth to availability of free amino acids. These results may have important implications for the use of mTOR inhibitors as therapeutics.


Subject(s)
Embryo, Mammalian/cytology , Multiprotein Complexes/metabolism , Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Albumins/metabolism , Amino Acids/metabolism , Animals , Cell Proliferation , Cell Survival , Eukaryota/classification , Eukaryota/cytology , Eukaryota/metabolism , Fibroblasts/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Pinocytosis , Proteins/chemistry , ras Proteins/metabolism
4.
Cell ; 157(2): 313-328, 2014 04 10.
Article in English | MEDLINE | ID: mdl-24656405

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Piperidines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Heterografts , Humans , Hydroxyquinolines/pharmacology , MAP Kinase Kinase 4/metabolism , Mice , Neoplasm Transplantation , Pinocytosis/drug effects , Vacuoles/metabolism , Zebrafish
5.
Genes Dev ; 34(19-20): 1253-1255, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004484

ABSTRACT

Cancer cells must adapt metabolism to thrive despite nutrient limitations in the tumor microenvironment. In this issue of Genes & Development, King and colleagues (pp. 1345-1358) report a role for transcriptional regulators of the Hippo pathway to facilitate protein scavenging and support proliferation under some nutrient-deprived conditions.


Subject(s)
Protein Serine-Threonine Kinases , Signal Transduction , Nutrients , Pinocytosis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation
6.
Genes Dev ; 34(19-20): 1345-1358, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32912902

ABSTRACT

The uptake of macromolecules and cellular debris through macropinocytosis has emerged as an important nutrient acquisition strategy of cancer cells. Genetic alterations commonly found in human cancers (e.g. mutations in KRAS or loss of PTEN) have been shown to increase macropinocytosis. To identify additional effectors that enable cell growth dependent on the uptake of extracellular proteins, pancreatic ductal adenocarcinoma (PDA) cells were selected for growth in medium where extracellular albumin was the obligate source of the essential amino acid leucine. Analysis of global changes in chromatin availability and gene expression revealed that PDA cells selected under these conditions exhibited elevated activity of the transcriptional activators Yap/Taz. Knockout of Yap/Taz prevented growth of PDA cells in leucine-deficient medium, but not in complete medium. Furthermore, constitutively active forms of Yap or Taz were sufficient to stimulate macropinocytosis of extracellular protein. In addition to promoting the uptake of plasma proteins, Yap/Taz also promoted the scavenging of apoptotic cell bodies and necrotic debris by PDA cells. The Yap/Taz transcriptional target Axl was found to be essential for cell growth dependent on the uptake of dead cells and cell debris. Together, these studies suggest that the Hippo pathway effectors Yap and Taz are important transcriptional regulators of endocytic nutrient uptake.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nutrients/metabolism , Pinocytosis/physiology , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Extracellular Space/metabolism , Humans , Mice , YAP-Signaling Proteins
7.
Proc Natl Acad Sci U S A ; 121(25): e2315481121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870060

ABSTRACT

Intracellular bacterial pathogens divert multiple cellular pathways to establish their niche and persist inside their host. Coxiella burnetii, the causative agent of Q fever, secretes bacterial effector proteins via its Type 4 secretion system to generate a Coxiella-containing vacuole (CCV). Manipulation of lipid and protein trafficking by these effectors is essential for bacterial replication and virulence. Here, we have characterized the lipid composition of CCVs and found that the effector Vice interacts with phosphoinositides and membranes enriched in phosphatidylserine and lysobisphosphatidic acid. Remarkably, eukaryotic cells ectopically expressing Vice present compartments that resemble early CCVs in both morphology and composition. We found that the biogenesis of these compartments relies on the double function of Vice. The effector protein initially localizes at the plasma membrane of eukaryotic cells where it triggers the internalization of large vacuoles by macropinocytosis. Then, Vice stabilizes these compartments by perturbing the ESCRT machinery. Collectively, our results reveal that Vice is an essential C. burnetii effector protein capable of hijacking two major cellular pathways to shape the bacterial replicative niche.


Subject(s)
Bacterial Proteins , Coxiella burnetii , Endosomal Sorting Complexes Required for Transport , Pinocytosis , Vacuoles , Endosomal Sorting Complexes Required for Transport/metabolism , Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , Humans , HeLa Cells , Cell Membrane/metabolism , Animals , Phosphatidylinositols/metabolism
8.
Annu Rev Biochem ; 79: 803-33, 2010.
Article in English | MEDLINE | ID: mdl-20196649

ABSTRACT

Although viruses are simple in structure and composition, their interactions with host cells are complex. Merely to gain entry, animal viruses make use of a repertoire of cellular processes that involve hundreds of cellular proteins. Although some viruses have the capacity to penetrate into the cytosol directly through the plasma membrane, most depend on endocytic uptake, vesicular transport through the cytoplasm, and delivery to endosomes and other intracellular organelles. The internalization may involve clathrin-mediated endocytosis (CME), macropinocytosis, caveolar/lipid raft-mediated endocytosis, or a variety of other still poorly characterized mechanisms. This review focuses on the cell biology of virus entry and the different strategies and endocytic mechanisms used by animal viruses.


Subject(s)
Endocytosis , Virus Internalization , Animals , Caveolae/metabolism , Clathrin/metabolism , Membrane Microdomains/metabolism , Phagocytosis , Pinocytosis
9.
Genes Dev ; 32(17-18): 1201-1214, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30143526

ABSTRACT

The architectural and biochemical features of the plasma membrane are governed by its intimate association with the underlying cortical cytoskeleton. The neurofibromatosis type 2 (NF2) tumor suppressor merlin and closely related membrane:cytoskeleton-linking protein ezrin organize the membrane:cytoskeleton interface, a critical cellular compartment that both regulates and is regulated by growth factor receptors. An example of this poorly understood interrelationship is macropinocytosis, an ancient process of nutrient uptake and membrane remodeling that can both be triggered by growth factors and manage receptor availability. We show that merlin deficiency primes the membrane:cytoskeleton interface for epidermal growth factor (EGF)-induced macropinocytosis via a mechanism involving increased cortical ezrin, altered actomyosin, and stabilized cholesterol-rich membranes. These changes profoundly alter EGF receptor (EGFR) trafficking in merlin-deficient cells, favoring increased membrane levels of its heterodimerization partner, ErbB2; clathrin-independent internalization; and recycling. Our work suggests that, unlike Ras transformed cells, merlin-deficient cells do not depend on macropinocytic protein scavenging and instead exploit macropinocytosis for receptor recycling. Finally, we provide evidence that the macropinocytic proficiency of NF2-deficient cells can be used for therapeutic uptake. This work provides new insight into fundamental mechanisms of macropinocytic uptake and processing and suggests new ways to interfere with or exploit macropinocytosis in NF2 mutant and other tumors.


Subject(s)
Cell Membrane/metabolism , Epidermal Growth Factor/physiology , ErbB Receptors/metabolism , Neurofibromin 2/physiology , Pinocytosis , Actomyosin/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Humans , Mice , Neurofibromin 2/genetics , Protein Biosynthesis
10.
Genes Cells ; 29(6): 512-520, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38597132

ABSTRACT

Macropinocytosis (MPC) is a large-scale endocytosis pathway that involves actin-dependent membrane ruffle formation and subsequent ruffle closure to generate macropinosomes for the uptake of fluid-phase cargos. MPC is categorized into two types: constitutive and stimuli-induced. Constitutive MPC in macrophages relies on extracellular Ca2+ sensing by a calcium-sensing receptor. However, the link between stimuli-induced MPC and Ca2+ remains unclear. Here, we find that both intracellular and extracellular Ca2+ are required for epidermal growth factor (EGF)-induced MPC in A431 human epidermoid carcinoma cells. Through investigation of mammalian homologs of coelomocyte uptake defective (CUP) genes, we identify ATP2B4, encoding for a Ca2+ pump called the plasma membrane calcium ATPase 4 (PMCA4), as a Ca2+-related regulator of EGF-induced MPC. Knockout (KO) of ATP2B4, as well as depletion of extracellular/intracellular Ca2+, inhibited ruffle closure and macropinosome formation, without affecting ruffle formation. We demonstrate the importance of PMCA4 activity itself, independent of interactions with other proteins via its C-terminus known as a PDZ domain-binding motif. Additionally, we show that ATP2B4-KO reduces EGF-stimulated Ca2+ oscillation during MPC. Our findings suggest that EGF-induced MPC requires ATP2B4-dependent Ca2+ dynamics.


Subject(s)
Calcium , Epidermal Growth Factor , Pinocytosis , Plasma Membrane Calcium-Transporting ATPases , Humans , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Calcium/metabolism , Cell Line, Tumor
11.
Nature ; 576(7787): 477-481, 2019 12.
Article in English | MEDLINE | ID: mdl-31827278

ABSTRACT

Oncogenic activation of RAS is associated with the acquisition of a unique set of metabolic dependencies that contribute to tumour cell fitness. Cells that express oncogenic RAS are able to internalize and degrade extracellular protein via a fluid-phase uptake mechanism termed macropinocytosis1. There is increasing recognition of the role of this RAS-dependent process in the generation of free amino acids that can be used to support tumour cell growth under nutrient-limiting conditions2. However, little is known about the molecular steps that mediate the induction of macropinocytosis by oncogenic RAS. Here we identify vacuolar ATPase (V-ATPase) as an essential regulator of RAS-induced macropinocytosis. Oncogenic RAS promotes the translocation of V-ATPase from intracellular membranes to the plasma membrane via a pathway that requires the activation of protein kinase A by a bicarbonate-dependent soluble adenylate cyclase. Accumulation of V-ATPase at the plasma membrane is necessary for the cholesterol-dependent plasma-membrane association of RAC1, a prerequisite for the stimulation of membrane ruffling and macropinocytosis. These observations establish a link between V-ATPase trafficking and nutrient supply by macropinocytosis that could be exploited to curtail the metabolic adaptation capacity of RAS-mutant tumour cells.


Subject(s)
Cell Membrane/enzymology , Oncogene Protein p21(ras)/metabolism , Pinocytosis , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Bicarbonates/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Sodium-Bicarbonate Symporters/metabolism
12.
Nature ; 568(7752): 410-414, 2019 04.
Article in English | MEDLINE | ID: mdl-30918400

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains recalcitrant to all forms of cancer treatment and carries a five-year survival rate of only 8%1. Inhibition of oncogenic KRAS (hereafter KRAS*), the earliest lesion in disease development that is present in more than 90% of PDACs, and its signalling surrogates has yielded encouraging preclinical results with experimental agents2-4. However, KRAS*-independent disease recurrence following genetic extinction of Kras* in mouse models anticipates the need for co-extinction strategies5,6. Multiple oncogenic processes are initiated at the cell surface, where KRAS* physically and functionally interacts to direct signalling that is essential for malignant transformation and tumour maintenance. Insights into the complexity of the functional cell-surface-protein repertoire (surfaceome) have been technologically limited until recently and-in the case of PDAC-the genetic control of the function and composition of the PDAC surfaceome in the context of KRAS* signalling remains largely unknown. Here we develop an unbiased, functional target-discovery platform to query KRAS*-dependent changes of the PDAC surfaceome, which reveals syndecan 1 (SDC1, also known as CD138) as a protein that is upregulated at the cell surface by KRAS*. Localization of SDC1 at the cell surface-where it regulates macropinocytosis, an essential metabolic pathway that fuels PDAC cell growth-is essential for disease maintenance and progression. Thus, our study forges a mechanistic link between KRAS* signalling and a targetable molecule driving nutrient salvage pathways in PDAC and validates oncogene-driven surfaceome annotation as a strategy to identify cancer-specific vulnerabilities.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Pinocytosis , Syndecan-1/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Disease Progression , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Male , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
13.
Mol Cell ; 67(6): 936-946.e5, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28918901

ABSTRACT

Scavenging of extracellular protein via macropinocytosis is an alternative to monomeric amino acid uptake. In pancreatic cancer, macropinocytosis is driven by oncogenic Ras signaling and contributes substantially to amino acid supply. While Ras signaling promotes scavenging, mTOR signaling suppresses it. Here, we present an integrated experimental-computational method that enables quantitative comparison of protein scavenging rates across cell lines and conditions. Using it, we find that, independently of mTORC1, amino acid scarcity induces protein scavenging and that under such conditions the impact of mTOR signaling on protein scavenging rate is minimal. Nevertheless, mTOR inhibition promotes growth of cells reliant on eating extracellular protein. This growth enhancement depends on mTORC1's canonical function in controlling translation rate: mTOR inhibition slows translation, thereby matching protein synthesis to the limited amino acid supply. Thus, paradoxically, in amino acid-poor conditions the pro-anabolic effects of mTORC1 are functionally opposed to growth.


Subject(s)
Amino Acids/metabolism , Energy Metabolism/drug effects , Fibroblasts/drug effects , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acids/deficiency , Animals , Cell Line , Cell Proliferation/drug effects , Computer Simulation , Fibroblasts/enzymology , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Mutation , Pinocytosis/drug effects , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection
14.
Proc Natl Acad Sci U S A ; 119(20): e2123261119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35561222

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) senses amino acids to control cell growth, metabolism, and autophagy. Some amino acids signal to mTORC1 through the Rag GTPase, whereas glutamine and asparagine activate mTORC1 through a Rag GTPase-independent pathway. Here, we show that the lysosomal glutamine and asparagine transporter SNAT7 activates mTORC1 after extracellular protein, such as albumin, is macropinocytosed. The N terminus of SNAT7 forms nutrient-sensitive interaction with mTORC1 and regulates mTORC1 activation independently of the Rag GTPases. Depletion of SNAT7 inhibits albumin-induced mTORC1 lysosomal localization and subsequent activation. Moreover, SNAT7 is essential to sustain KRAS-driven pancreatic cancer cell growth through mTORC1. Thus, SNAT7 links glutamine and asparagine signaling from extracellular protein to mTORC1 independently of the Rag GTPases and is required for macropinocytosis-mediated mTORC1 activation and pancreatic cancer cell growth.


Subject(s)
Amino Acid Transport Systems, Neutral , Lysosomes , Mechanistic Target of Rapamycin Complex 1 , Pinocytosis , Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Asparagine/metabolism , Glutamine/metabolism , Humans , Lysosomes/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction
15.
Annu Rev Biochem ; 78: 857-902, 2009.
Article in English | MEDLINE | ID: mdl-19317650

ABSTRACT

Endocytic mechanisms control the lipid and protein composition of the plasma membrane, thereby regulating how cells interact with their environments. Here, we review what is known about mammalian endocytic mechanisms, with focus on the cellular proteins that control these events. We discuss the well-studied clathrin-mediated endocytic mechanisms and dissect endocytic pathways that proceed independently of clathrin. These clathrin-independent pathways include the CLIC/GEEC endocytic pathway, arf6-dependent endocytosis, flotillin-dependent endocytosis, macropinocytosis, circular doral ruffles, phagocytosis, and trans-endocytosis. We also critically review the role of caveolae and caveolin1 in endocytosis. We highlight the roles of lipids, membrane curvature-modulating proteins, small G proteins, actin, and dynamin in endocytic pathways. We discuss the functional relevance of distinct endocytic pathways and emphasize the importance of studying these pathways to understand human disease processes.


Subject(s)
Endocytosis , Animals , Caveolae/metabolism , Clathrin/metabolism , Humans , Phagocytosis , Pinocytosis , Protein Transport
16.
Trends Biochem Sci ; 45(6): 459-461, 2020 06.
Article in English | MEDLINE | ID: mdl-32413322

ABSTRACT

The RAS oncoprotein drives elevated macropinocytosis, a metabolic process essential for cancer growth. A recent study by Ramirez et al. elucidated a mechanism whereby RAS controls V-ATPase association with the plasma membrane to drive RAC1 GTPase-dependent macropinocytosis. Potentially actionable targets to disrupt this RAS-dependent nutrient acquisition pathway were identified.


Subject(s)
Binge Drinking , Neoplasms , Adenosine Triphosphatases , Cell Membrane , Humans , Neoplasms/genetics , Pinocytosis
17.
J Cell Mol Med ; 28(11): e18477, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38853458

ABSTRACT

Given the pathological role of Tau aggregation in Alzheimer's disease (AD), our laboratory previously developed the novel Tau aggregation inhibitor peptide, RI-AG03. As Tau aggregates accumulate intracellularly, it is essential that the peptide can traverse the cell membrane. Here we examine the cellular uptake and intracellular trafficking of RI-AG03, in both a free and liposome-conjugated form. We also characterize the impact of adding the cell-penetrating peptide (CPP) sequences, polyarginine (polyR) or transactivator of transcription (TAT), to RI-AG03. Our data show that liposome conjugation of CPP containing RI-AG03 peptides, with either the polyR or TAT sequence, increased cellular liposome association three-fold. Inhibition of macropinocytosis modestly reduced the uptake of unconjugated and RI-AG03-polyR-linked liposomes, while having no effect on RI-AG03-TAT-conjugated liposome uptake. Further supporting macropinocytosis-mediated internalization, a 'fair' co-localisation of the free and liposome-conjugated RI-AG03-polyR peptide with macropinosomes and lysosomes was observed. Interestingly, we also demonstrate that RI-AG03-polyR detaches from liposomes following cellular uptake, thereby largely evading organellar entrapment. Collectively, our data indicate that direct membrane penetration and macropinocytosis are key routes for the internalization of liposomes conjugated with CPP containing RI-AG03. Our study also demonstrates that peptide-liposomes are suitable nanocarriers for the cellular delivery of RI-AG03, furthering their potential use in targeting Tau pathology in AD.


Subject(s)
Cell-Penetrating Peptides , Liposomes , Nanoparticles , Pinocytosis , tau Proteins , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Liposomes/chemistry , Humans , tau Proteins/metabolism , tau Proteins/chemistry , Nanoparticles/chemistry , Pinocytosis/drug effects , Peptides/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Lysosomes/metabolism , Drug Delivery Systems/methods
18.
J Biol Chem ; 299(4): 104605, 2023 04.
Article in English | MEDLINE | ID: mdl-36918100

ABSTRACT

Pseudorabies virus (PRV) has become a "new life-threatening zoonosis" since the human-originated PRV strain was first isolated in 2020. To identify novel anti-PRV agents, we screened a total of 107 ß-carboline derivatives and found 20 compounds displaying antiviral activity against PRV. Among them, 14 compounds showed better antiviral activity than acyclovir. We found that compound 45 exhibited the strongest anti-PRV activity with an IC50 value of less than 40 nM. Our in vivo studies showed that treatment with 45 significantly reduced the viral loads and protected mice challenged with PRV. To clarify the mode of action of 45, we conducted a time of addition assay, an adsorption assay, and an entry assay. Our results indicated that 45 neither had a virucidal effect nor affected viral adsorption while significantly inhibiting PRV entry. Using the FITC-dextran uptake assay, we determined that 45 inhibits macropinocytosis. The actin-dependent plasma membrane protrusion, which is important for macropinocytosis, was also suppressed by 45. Furthermore, the kinase DYRK1A (dual-specificity tyrosine phosphorylation-regulated kinase 1A) was predicted to be a potential target for 45. The binding of 45 to DYRK1A was confirmed by drug affinity responsive target stability and cellular thermal shift assay. Further analysis revealed that knockdown of DYRK1A by siRNA suppressed PRV macropinocytosis and the tumor necrosis factor alpha-TNF-induced formation of protrusions. These results suggested that 45 could restrain PRV macropinocytosis by targeting DYRK1A. Together, these findings reveal a unique mechanism through which ß-carboline derivatives restrain PRV infection, pointing to their potential value in the development of anti-PRV agents.


Subject(s)
Antiviral Agents , Carbolines , Herpesvirus 1, Suid , Animals , Humans , Mice , Acyclovir/pharmacology , Acyclovir/toxicity , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Carbolines/chemistry , Carbolines/pharmacology , Carbolines/therapeutic use , Gene Knockdown Techniques , Herpesvirus 1, Suid/drug effects , Inhibitory Concentration 50 , Pinocytosis/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pseudorabies/drug therapy , Pseudorabies/prevention & control , Pseudorabies/virology , Virus Internalization/drug effects , HeLa Cells , Models, Chemical , Dyrk Kinases
19.
EMBO J ; 39(20): e104862, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32853409

ABSTRACT

Genetic variation in LRRK2 associates with the susceptibility to Parkinson's disease, Crohn's disease, and mycobacteria infection. High expression of LRRK2 and its substrate Rab10 occurs in phagocytic cells in the immune system. In mouse and human primary macrophages, dendritic cells, and microglia-like cells, we find that Rab10 specifically regulates a specialized form of endocytosis known as macropinocytosis, without affecting phagocytosis or clathrin-mediated endocytosis. LRRK2 phosphorylates cytoplasmic PI(3,4,5)P3-positive GTP-Rab10, before EEA1 and Rab5 recruitment to early macropinosomes occurs. Macropinosome cargo in macrophages includes CCR5, CD11b, and MHCII, and LRRK2-phosphorylation of Rab10 potently blocks EHBP1L1-mediated recycling tubules and cargo turnover. EHBP1L1 overexpression competitively inhibits LRRK2-phosphorylation of Rab10, mimicking the effects of LRRK2 kinase inhibition in promoting cargo recycling. Both Rab10 knockdown and LRRK2 kinase inhibition potently suppress the maturation of macropinosome-derived CCR5-loaded signaling endosomes that are critical for CCL5-induced immunological responses that include Akt activation and chemotaxis. These data support a novel signaling axis in the endolysosomal system whereby LRRK2-mediated Rab10 phosphorylation stalls vesicle fast recycling to promote PI3K-Akt immunological responses.


Subject(s)
Carrier Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Macrophages/metabolism , Phagocytes/immunology , Pinocytosis/genetics , rab GTP-Binding Proteins/metabolism , Animals , Cell Membrane/metabolism , Chemokine CCL5/pharmacology , Chemotaxis/genetics , Dendritic Cells/metabolism , Endosomes/drug effects , Endosomes/metabolism , Female , Gene Knockdown Techniques , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Macrophages/drug effects , Male , Mass Spectrometry , Mice , Mice, Transgenic , Microglia/metabolism , Monocytes/drug effects , Monocytes/metabolism , Mutation , Phagocytes/drug effects , Phagocytes/metabolism , Phosphorylation , Pinocytosis/drug effects , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , rab GTP-Binding Proteins/genetics
20.
J Cell Sci ; 135(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35267018

ABSTRACT

Macropinocytosis mediates non-selective bulk uptake of extracellular fluid. It is the major route by which axenic Dictyostelium cells obtain nutrients and has emerged as a nutrient-scavenging pathway in mammalian cells. How environmental and cellular nutrient status modulates macropinocytic activity is not well understood. By developing a high-content imaging-based genetic screen in Dictyostelium discoideum we identified Slc15A, an oligopeptide transporter located at the plasma membrane and early macropinosome, as a novel macropinocytosis regulator. We show that deletion of slc15A but not two other related slc15 genes, leads to reduced macropinocytosis, reduced cell growth and aberrantly increased autophagy in cells grown in nutrient-rich medium. Expression of Slc15A protein or supplying cells with free amino acids rescues these defects. In contrast, expression of transport-defective Slc15A or supplying cells with amino acids in their di-peptide forms fails to rescue these defects. Therefore, Slc15A modulates the level of macropinocytosis by maintaining the intracellular availability of key amino acids through extraction of oligopeptides from the early macropinocytic pathway. We propose that Slc15A constitutes part of a positive feedback mechanism coupling cellular nutrient status and macropinocytosis. This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Dictyostelium , Animals , Dictyostelium/genetics , Endosomes , Humans , Mammals , Nutrients , Oligopeptides , Pinocytosis
SELECTION OF CITATIONS
SEARCH DETAIL