Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 572
1.
J Cell Mol Med ; 28(11): e18477, 2024 Jun.
Article En | MEDLINE | ID: mdl-38853458

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.


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
2.
Anticancer Res ; 44(5): 1939-1946, 2024 May.
Article En | MEDLINE | ID: mdl-38677763

BACKGROUND/AIM: Macropinocytosis is a non-selective form of endocytosis that facilitates the uptake of extracellular substances, such as nutrients and macromolecules, into the cells. In KRAS-driven cancers, including pancreatic ductal adenocarcinoma, macropinocytosis and subsequent lysosomal utilization are known to be enhanced to overcome metabolic stress. In this study, we investigated the role of Casein Kinase 2 (CK2) inhibition in macropinocytosis and subsequent metabolic processes in KRAS mutant cholangiocarcinoma (CCA) cell lines. MATERIALS AND METHODS: The bovine serum albumin (BSA) uptake indicating macropinocytosis was performed by flow cytometry using the HuCCT1 KRAS mutant CCA cell line. To validate macropinosome, the Rab7 and LAMP2 were labeled and analyzed via immunocytochemistry and western blot. The CX-4945 (Silmitasertib), CK2 inhibitor, was used to investigate the role of CK2 in macropinocytosis and subsequent lysosomal metabolism. RESULTS: The TFK-1, a KRAS wild-type CCA cell line, showed only apoptotic morphological changes. However, the HuCCT1 cell line showed macropinocytosis. Although CX-4945 induced morphological changes accompanied by the accumulation of intracellular vacuoles and cell death, the level of macropinocytosis did not change. These intracellular vacuoles were identified as late macropinosomes, representing Rab7+ vesicles before fusion with lysosomes. In addition, CX-4945 suppressed LAMP2 expression following the inhibition of the Akt-mTOR signaling pathway, which interrupts mature macropinosome and lysosomal metabolic utilization. CONCLUSION: Macropinocytosis is used as an energy source in the KRAS mutant CCA cell line HuCCT1. The inhibition of CK2 by CX-4945 leads to cell death in HuCCT1 cells through alteration of the lysosome-dependent metabolism.


Bile Duct Neoplasms , Casein Kinase II , Cholangiocarcinoma , Lysosomes , Mutation , Naphthyridines , Phenazines , Pinocytosis , Piperazines , Proto-Oncogene Proteins p21(ras) , Humans , Lysosomes/metabolism , Cell Line, Tumor , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Pinocytosis/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Casein Kinase II/metabolism , Casein Kinase II/genetics , Casein Kinase II/antagonists & inhibitors , Piperazines/pharmacology , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , rab7 GTP-Binding Proteins/metabolism , Cell Death/drug effects , Apoptosis/drug effects , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics
3.
J Biol Chem ; 299(4): 104605, 2023 04.
Article En | MEDLINE | ID: mdl-36918100

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.


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
4.
Sci Rep ; 12(1): 6322, 2022 04 15.
Article En | MEDLINE | ID: mdl-35428847

Macropinocytosis is a type of endocytosis accompanied by actin rearrangement-driven membrane deformation, such as lamellipodia formation and membrane ruffling, followed by the formation of large vesicles, macropinosomes. Ras-transformed cancer cells efficiently acquire exogenous amino acids for their survival through macropinocytosis. Thus, inhibition of macropinocytosis is a promising strategy for cancer therapy. To date, few specific agents that inhibit macropinocytosis have been developed. Here, focusing on the mechanosensitive ion channel Piezo1, we found that Yoda1, a Piezo1 agonist, potently inhibits macropinocytosis induced by epidermal growth factor (EGF). The inhibition of ruffle formation by Yoda1 was dependent on the extracellular Ca2+ influx through Piezo1 and on the activation of the calcium-activated potassium channel KCa3.1. This suggests that Ca2+ ions can regulate EGF-stimulated macropinocytosis. We propose the potential for macropinocytosis inhibition through the regulation of a mechanosensitive channel activity using chemical tools.


Carcinoma, Squamous Cell , Epidermal Growth Factor , Ion Channels , Pyrazines , Thiadiazoles , Biological Transport , Calcium/metabolism , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Humans , Ion Channels/agonists , Ion Channels/metabolism , Pinocytosis/drug effects
5.
J Exp Clin Cancer Res ; 41(1): 98, 2022 Mar 14.
Article En | MEDLINE | ID: mdl-35287706

BACKGROUND: Macropinocytosis, an important nutrient-scavenging pathway in certain cancer cells, allows cells to compensate for intracellular amino acid deficiency under nutrient-poor conditions. Ferroptosis caused by cysteine depletion plays a pivotal role in sorafenib responses during hepatocellular carcinoma (HCC) therapy. However, it is not known whether macropinocytosis functions as an alternative pathway to acquire cysteine in sorafenib-treated HCC, and whether it subsequently mitigates sorafenib-induced ferroptosis. This study aimed to investigate whether sorafenib drives macropinocytosis induction, and how macropinocytosis confers ferroptosis resistance on HCC cells. METHODS: Macropinocytosis, both in HCC cells and HCC tissues, was evaluated by measuring TMR-dextran uptake or lysosomal degradation of DQ-BSA, and ferroptosis was evaluated via C11-BODIPY fluorescence and 4-HNE staining. Sorafenib-induced ferroptosis and macropinocytosis were validated in tumor tissues taken from HCC patients who underwent ultrasound-guided needle biopsy. RESULTS: Sorafenib increased macropinocytosis in human HCC specimens and xenografted HCC tissues. Sorafenib-induced mitochondrial dysfunction was responsible for activation of PI3K-RAC1-PAK1 signaling, and amplified macropinocytosis in HCC. Importantly, macropinocytosis prevented sorafenib-induced ferroptosis by replenishing intracellular cysteine that was depleted by sorafenib treatment; this rendered HCC cells resistant to sorafenib. Finally, inhibition of macropinocytosis by amiloride markedly enhanced the anti-tumor effect of sorafenib, and sensitized resistant tumors to sorafenib. CONCLUSION: In summary, sorafenib induced macropinocytosis, which conferred drug resistance by mitigating sorafenib-induced ferroptosis. Thus, targeting macropinocytosis is a promising therapeutic strategy to facilitate ferroptosis-based therapy for HCC.


Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/drug therapy , Cysteine/therapeutic use , Ferroptosis/drug effects , Liver Neoplasms/complications , Liver Neoplasms/drug therapy , Pinocytosis/drug effects , Protein Kinase Inhibitors/therapeutic use , Sorafenib/therapeutic use , Animals , Carcinoma, Hepatocellular/pathology , Cysteine/pharmacology , Female , Humans , Liver Neoplasms/pathology , Male , Mice , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology
6.
J Cell Sci ; 135(4)2022 02 15.
Article En | MEDLINE | ID: mdl-35107133

The internalization of solutes by macropinocytosis provides an essential route for nutrient uptake in many cells. Macrophages increase macropinocytosis in response to growth factors and other stimuli. To test the hypothesis that nutrient environments modulate solute uptake by macropinocytosis, this study analyzed the effects of extracellular amino acids on the accumulation of fluorescent fluid-phase probes in murine macrophages. Nine amino acids, added individually or together, were capable of suppressing macropinocytosis in murine bone marrow-derived macrophages stimulated with the growth factors colony stimulating factor 1 (CSF1) or interleukin 34, both ligands of the CSF1 receptor (CSF1R). The suppressive amino acids did not inhibit macropinocytosis in response to lipopolysaccharide, the chemokine CXCL12, or the tumor promoter phorbol myristate acetate. Suppressive amino acids promoted release of CSF1R from cells and resulted in the formation of smaller macropinosomes in response to CSF1. This suppression of growth factor-stimulated macropinocytosis indicates that different nutrient environments modulate CSF1R levels and bulk ingestion by macropinocytosis, with likely consequences for macrophage growth and function.


Amino Acids , Macrophage Colony-Stimulating Factor , Animals , Endosomes/metabolism , Macrophages/metabolism , Mice , Pinocytosis/drug effects , Receptor Protein-Tyrosine Kinases/metabolism
7.
Nat Commun ; 13(1): 954, 2022 02 17.
Article En | MEDLINE | ID: mdl-35177645

Hepatocellular carcinoma (HCC) invariably exhibits inadequate O2 (hypoxia) and nutrient supply. Hypoxia-inducible factor (HIF) mediates cascades of molecular events that enable cancer cells to adapt and propagate. Macropinocytosis is an endocytic process initiated by membrane ruffling, causing the engulfment of extracellular fluids (proteins), protein digestion and subsequent incorporation into the biomass. We show that macropinocytosis occurs universally in HCC under hypoxia. HIF-1 activates the transcription of a membrane ruffling protein, EH domain-containing protein 2 (EHD2), to initiate macropinocytosis. Knockout of HIF-1 or EHD2 represses hypoxia-induced macropinocytosis and prevents hypoxic HCC cells from scavenging protein that support cell growth. Germline or somatic deletion of Ehd2 suppresses macropinocytosis and HCC development in mice. Intriguingly, EHD2 is overexpressed in HCC. Consistently, HIF-1 or macropinocytosis inhibitor suppresses macropinocytosis and HCC development. Thus, we show that hypoxia induces macropinocytosis through the HIF/EHD2 pathway in HCC cells, harnessing extracellular protein as a nutrient to survive.


Carcinoma, Hepatocellular/immunology , Carrier Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/immunology , Pinocytosis/immunology , Tumor Hypoxia/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/immunology , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Knockout , Pinocytosis/drug effects , Pinocytosis/genetics , Proof of Concept Study , Tumor Hypoxia/immunology , Xenograft Model Antitumor Assays
8.
Cancer Sci ; 113(2): 609-621, 2022 Feb.
Article En | MEDLINE | ID: mdl-34874595

Various cell types secrete exosomes into their surrounding extracellular space, which consequently affect the function and activity of recipient cells. Numerous studies have showed that tumor cell-derived exosomes play important roles in tumor growth and progression. Although a variety of endocytic pathways are reportedly involved in the cellular uptake of exosomes, detailed mechanisms remain unknown. The present study demonstrated that treatment with recombinant epidermal growth factor (EGF) time- and dose-dependently promoted cellular uptake of oral squamous cell carcinoma (OSCC) cell-derived exosomes into OSCC cells themselves. Conversely, EGF receptor (EGFR) knockdown and treatment with EGFR inhibitors, including erlotinib and cetuximab, abrogated OSCC cell uptake of exosomes. The macropinocytosis inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) blocked the effects of active EGF/EGFR signaling on uptake of OSCC cell-derived exosomes. These EGFR inhibitors also suppressed OSCC cell-derived exosome-induced proliferation, migration, invasion, stemness, and chemoresistance of OSCC cells. Taken together, the data presented herein suggest that EGFR inhibitors might inhibit the malignant potential of OSCC cells through direct inhibition of not only EGFR downstream signaling pathway but also cellular uptake of OSCC cell-derived exosomes through macropinocytosis.


Epidermal Growth Factor/metabolism , Exosomes/metabolism , Mouth Neoplasms/metabolism , Pinocytosis , Squamous Cell Carcinoma of Head and Neck/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mouth Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pinocytosis/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Drug Deliv ; 28(1): 2480-2494, 2021 Dec.
Article En | MEDLINE | ID: mdl-34766543

Cisplatin (DDP) is a first-line chemotherapeutic drug applied for the treatment of oral squamous cell carcinoma (OSCC). The anticancer activity of DDP is tightly linked to its intracellular uptake. It is unwise to increase the DDP intake by increasing the dose or shortening the dosing interval because of the severe systemic toxicity (nephrotoxicity, ototoxicity and neurotoxicity) in DDP application. The main uptake pathways of DDP include passive diffusion and active transporter transport. Therefore, finding additional uptake pathways that can improve the effective intracellular concentration of DDP is critical. Macropinocytosis, an endocytic mechanism for extracellular material absorption, contributes to the intracellular uptake of anticancer drugs. No research has been conducted to determine whether macropinocytosis can augment the intracellular uptake of DDP in OSCC cells or not. Based on that, we proved for the first time that silmitasertib (previously CX-4945) could trigger macropinocytosis, which may increase the intracellular uptake of DDP and enhance apoptosis via in vivo and in vitro experiments. We hope that our findings will inspire a new approach for the application of DDP in cancer treatment.


Antineoplastic Agents/pharmacokinetics , Naphthyridines/pharmacology , Phenazines/pharmacology , Pinocytosis/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacokinetics , Drug Liberation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Mouth Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Int J Biol Macromol ; 190: 101-112, 2021 Nov 01.
Article En | MEDLINE | ID: mdl-34478790

The development of selenized polysaccharides is a promising strategy for the dietary selenium supplementation. The purpose of this research is to determine the influence of selenium on the structure and bioactivity of a polysaccharide fraction (MPN) isolated from Ganoderma lucidum mycelia. After biological selenium enrichment, the selenium content in the selenized polysaccharide (SeMPN) was 18.91 ± 1.8 µg/g. SeMPN had a slightly lower molecular weight than MPN, but the carbohydrate content and monosaccharide composition remained identical. Additionally, the band at 606 cm-1 in MPN changed to 615 cm-1 in SeMPN as revealed by FT-IR spectra. No significant changes were observed in the types and ratios of glycosidic linkages, as determined by NMR spectroscopy. Extracellular and intracellular antioxidant assays demonstrated that SeMPN was more effective than MPN in scavenging free radicals, inhibiting AAPH-induced erythrocyte hemolysis, and protecting catalase (CAT) and glutathione peroxidase (GSH-Px) activity in H2O2-injured PC12 cells. Additionally, SeMPN had a higher increase effect on RAW 264.7 cells's pinocytic and phagocytic capacity, as well as their production of NO, TNF-α, and IL-6. SeMPN could be as potential functional selenium supplementation.


Mycelium/chemistry , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , Reishi/chemistry , Selenium/chemistry , Animals , Antioxidants/pharmacology , Carbon-13 Magnetic Resonance Spectroscopy , Catalase/metabolism , Cell Survival/drug effects , Erythrocytes/drug effects , Erythrocytes/physiology , Glutathione Peroxidase/metabolism , Glycosides/chemistry , Hemolysis/drug effects , Interleukin-6/metabolism , Macrophages/drug effects , Mice , Molecular Weight , Monosaccharides/analysis , Nitric Oxide/biosynthesis , PC12 Cells , Phagocytosis/drug effects , Pinocytosis/drug effects , RAW 264.7 Cells , Rats , Reactive Oxygen Species/metabolism , Spectroscopy, Fourier Transform Infrared , Tumor Necrosis Factor-alpha/metabolism
11.
Nat Commun ; 12(1): 4838, 2021 08 10.
Article En | MEDLINE | ID: mdl-34376698

Macropinosomes are formed by shaping actin-rich plasma membrane ruffles into large intracellular organelles in a phosphatidylinositol 3-kinase (PI3K)-coordinated manner. Here, we utilize lattice lightsheet microscopy and image visualization methods to map the three-dimensional structure and dynamics of macropinosome formation relative to PI3K activity. We show that multiple ruffling morphologies produce macropinosomes and that the majority form through collisions of adjacent PI3K-rich ruffles. By combining multiple volumetric representations of the plasma membrane structure and PI3K products, we show that PI3K activity begins early throughout the entire ruffle volume and continues to increase until peak activity concentrates at the base of the ruffle after the macropinosome closes. Additionally, areas of the plasma membrane rich in ruffling had increased PI3K activity and produced many macropinosomes of various sizes. Pharmacologic inhibition of PI3K activity had little effect on the rate and morphology of membrane ruffling, demonstrating that early production of 3'-phosphoinositides within ruffles plays a minor role in regulating their morphology. However, 3'-phosphoinositides are critical for the fusogenic activity that seals ruffles into macropinosomes. Taken together, these data indicate that local PI3K activity is amplified in ruffles and serves as a priming mechanism for closure and sealing of ruffles into macropinosomes.


Cell Membrane/metabolism , Microscopy, Fluorescence/methods , Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis/physiology , Animals , Cell Membrane/drug effects , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Macrophages/cytology , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Microscopy, Electron, Scanning , Morpholines/pharmacology , Phosphatidylinositols/metabolism , Pinocytosis/drug effects , RAW 264.7 Cells
12.
Molecules ; 26(15)2021 Jul 30.
Article En | MEDLINE | ID: mdl-34361779

Delivering nucleic acids into the endothelium has great potential in treating vascular diseases. However, endothelial cells, which line the vasculature, are considered as sensitive in nature and hard to transfect. Low transfection efficacies in endothelial cells limit their potential therapeutic applications. Towards improving the transfection efficiency, we made an effort to understand the internalization of lipoplexes into the cells, which is the first and most critical step in nucleic acid transfections. In this study, we demonstrated that the transient modulation of caveolae/lipid rafts mediated endocytosis with the cholesterol-sequestrating agents, nystatin, filipin III, and siRNA against Cav-1, which significantly increased the transfection properties of cationic lipid-(2-hydroxy-N-methyl-N,N-bis(2-tetradecanamidoethyl)ethanaminium chloride), namely, amide liposomes in combination with 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) (AD Liposomes) in liver sinusoidal endothelial cells (SK-Hep1). In particular, nystatin was found to be highly effective with 2-3-fold enhanced transfection efficacy when compared with amide liposomes in combination with Cholesterol (AC), by switching lipoplex internalization predominantly through clathrin-mediated endocytosis and macropinocytosis.


Caveolae/drug effects , Cholesterol/chemistry , Endothelial Cells/drug effects , Liposomes/chemistry , Membrane Microdomains/drug effects , Transfection/methods , Animals , Caveolae/chemistry , Caveolae/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Transformed , Cholesterol/metabolism , Clathrin/metabolism , DNA/chemistry , DNA/metabolism , Endocytosis/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Filipin/chemistry , Filipin/pharmacology , Gene Expression , Liposomes/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Nystatin/chemistry , Nystatin/pharmacology , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacology , Pinocytosis/drug effects , Plasmids/chemistry , Plasmids/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats
13.
Int J Biol Macromol ; 188: 729-739, 2021 Oct 01.
Article En | MEDLINE | ID: mdl-34389393

A neutral polysaccharide designated as CMDP-1a (molecular mass 9.263 kDa) was isolated from Cucurbita moschata Duch through hot water extraction, ethanol precipitation, and column chromatography. On the basis of methylation, fourier-transform infrared, monosaccharide composition, and one- and two-dimensional nuclear magnetic resonance spectroscopy analyses, the structure of CMDP-1a was determined to be a backbone composed of α-1,4 linked glucopyranosyl residues with α-Glcp residue linkage at backbone C-6. Atomic force microscopy and scanning electron microscopy analyses revealed that CMDP-1a had a spherical conformation in solution. In immunostimulation assays, CMDP-1a promoted the proliferation of RAW 264.7 macrophages and significantly enhanced their pinocytic and phagocytic capacity. Furthermore, CMDP-1a induced the M1 polarization of original macrophages and the conversion of macrophages from M2 to M1, thereby modulating the balance of M1/M2 macrophages. These results indicated that CMDP-1a might be a potential immunomodulator for food purposes.


Cucurbita/chemistry , Immunologic Factors/pharmacology , Polysaccharides/pharmacology , Animals , Cell Polarity/drug effects , Cytokines/genetics , Cytokines/metabolism , Macrophages/drug effects , Macrophages/immunology , Magnetic Resonance Spectroscopy , Methylation , Mice , Models, Biological , Molecular Weight , Monosaccharides/analysis , Phagocytosis/drug effects , Pinocytosis/drug effects , Polysaccharides/isolation & purification , Polysaccharides/ultrastructure , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
14.
FASEB J ; 35(9): e21742, 2021 09.
Article En | MEDLINE | ID: mdl-34403506

Withdrawal from contact inhibition is necessary for epithelial cancer precursor cells to initiate cell growth and motility. Nevertheless, little is understood about the mechanism for the sudden initiation of cell growth under static conditions. We focused on cellular junctions as one region where breaking out of contact inhibition occurs. In well-differentiated endometrial cancer cells, Sawano, the ligand administration for tricellular tight junction protein LSR, which transiently decreased the robust junction property, caused an abrupt increase in cell motility and consequent excessive multilayered cell growth despite being under contact inhibition conditions. We observed that macropinocytosis essentially and temporarily occurred as an antecedent event for the above process at intercellular junctions without disruption of the junction apparatus but not at the apical plasma membrane. Collectively, we concluded that the formation of macropinocytosis, which is derived from tight junction-mediated signaling, was triggered for the initiation of cell growth in static precancerous epithelium.


Cell Adhesion , Contact Inhibition , Pinocytosis , Receptors, Lipoprotein/metabolism , Transcription Factors/metabolism , Bacterial Toxins/pharmacology , Binding Sites , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Phenotype , Pinocytosis/drug effects , Protein Transport , Vacuoles/drug effects , Vacuoles/metabolism , rac GTP-Binding Proteins/metabolism
15.
Carbohydr Polym ; 269: 118288, 2021 Oct 01.
Article En | MEDLINE | ID: mdl-34294314

A pectic polysaccharide (named CMDP-4b) with a molecular weight of 31.97 kDa was extracted from Cucurbita moschata Duch and purified by column chromatography. On the basis of methylation, Fourier-transform infrared, monosaccharide composition, and one- and two-dimensional nuclear magnetic resonance spectroscopy analyses, the structure of CMDP-4b was determined to be composed of an α-1,4-linked homogalacturonan backbone, which was slightly acetylated and highly methyl-esterified, and branched at the O-3 position of the →4)-α-D-GalpA-6-OMe-(1→. Immunomodulatory assays showed that CMDP-4b not only induced the secretion of nitrous oxide and cytokines (i.e. IL-1ß, TNF-α, and IL-6) but also promoted pinocytic and phagocytic activities of macrophages, suggesting that CMDP-4b possessed immunomodulatory activity. Moreover, toll-like receptor 4 and complement receptor 3 may play a critical role in CMDP-4b-induced macrophage activation through the NF-κB and the MAPKs signaling pathways. Our study provides the molecular basis for the potential use of CMDP-4b as a natural immunostimulant.


Cucurbita/chemistry , Immunologic Factors/pharmacology , Pectins/pharmacology , Animals , Cell Survival/drug effects , Cytokines/metabolism , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Pectins/chemistry , Pectins/isolation & purification , Phagocytosis/drug effects , Pinocytosis/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects
16.
Front Immunol ; 12: 649600, 2021.
Article En | MEDLINE | ID: mdl-34135890

Using the optogenetic photo-manipulation of photoactivatable (PA)-Rac1, remarkable cell surface ruffling and the formation of a macropinocytic cup (premacropinosome) could be induced in the region of RAW264 macrophages irradiated with blue light due to the activation of PA-Rac1. However, the completion of macropinosome formation did not occur until Rac1 was deactivated by the removal of the light stimulus. Following PA-Rac1 deactivation, some premacropinosomes closed into intracellular macropinosomes, whereas many others transformed into long Rab10-positive tubules without forming typical macropinosomes. These Rab10-positive tubules moved centripetally towards the perinuclear Golgi region along microtubules. Surprisingly, these Rab10-positive tubules did not contain any endosome/lysosome compartment markers, such as Rab5, Rab7, or LAMP1, suggesting that the Rab10-positive tubules were not part of the degradation pathway for lysosomes. These Rab10-positive tubules were distinct from recycling endosomal compartments, which are labeled with Rab4, Rab11, or SNX1. These findings suggested that these Rab10-positive tubules may be a part of non-degradative endocytic pathway that has never been known. The formation of Rab10-positive tubules from premacropinosomes was also observed in control and phorbol myristate acetate (PMA)-stimulated macrophages, although their frequencies were low. Interestingly, the formation of Rab10-positive premacropinosomes and tubules was not inhibited by phosphoinositide 3-kinase (PI3K) inhibitors, while the classical macropinosome formation requires PI3K activity. Thus, this study provides evidence to support the existence of Rab10-positive tubules as a novel endocytic pathway that diverges from canonical macropinocytosis.


Endocytosis/immunology , Macrophages/immunology , Microtubules/metabolism , Pinocytosis/immunology , rab GTP-Binding Proteins/metabolism , Animals , Endocytosis/drug effects , Endocytosis/radiation effects , Golgi Apparatus/metabolism , Intravital Microscopy , Light , Macrophages/drug effects , Macrophages/metabolism , Mice , Microtubules/immunology , Microtubules/radiation effects , Neuropeptides/genetics , Neuropeptides/metabolism , Optogenetics , Pinocytosis/drug effects , Pinocytosis/radiation effects , RAW 264.7 Cells , Tetradecanoylphorbol Acetate/pharmacology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
17.
JCI Insight ; 6(11)2021 06 08.
Article En | MEDLINE | ID: mdl-33914709

Patients with chronic kidney disease (CKD) and end-stage renal disease suffer from increased cardiovascular events and cardiac mortality. Prior studies have demonstrated that a portion of this enhanced risk can be attributed to the accumulation of microbiota-derived toxic metabolites, with most studies focusing on the sulfonated form of p-cresol (PCS). However, unconjugated p-cresol (uPC) itself was never assessed due to rapid and extensive first-pass metabolism that results in negligible serum concentrations of uPC. These reports thus failed to consider the host exposure to uPC prior to hepatic metabolism. In the current study, not only did we measure the effect of altering the intestinal microbiota on lipid accumulation in coronary arteries, but we also examined macrophage lipid uptake and handling pathways in response to uPC. We found that atherosclerosis-prone mice fed a high-fat diet exhibited significantly higher coronary artery lipid deposits upon receiving fecal material from CKD mice. Furthermore, treatment with uPC increased total cholesterol, triglycerides, and hepatic and aortic fatty deposits in non-CKD mice. Studies employing an in vitro macrophage model demonstrated that uPC exposure increased apoptosis whereas PCS did not. Additionally, uPC exhibited higher potency than PCS to stimulate LDL uptake and only uPC induced endocytosis- and pinocytosis-related genes. Pharmacological inhibition of varying cholesterol influx and efflux systems indicated that uPC increased macrophage LDL uptake by activating macropinocytosis. Overall, these findings indicate that uPC itself had a distinct effect on macrophage biology that might have contributed to increased cardiovascular risk in patients with CKD.


Aorta/metabolism , Cholesterol, LDL/metabolism , Coronary Artery Disease/metabolism , Cresols/metabolism , Gastrointestinal Microbiome , Liver/metabolism , Macrophages/metabolism , Pinocytosis/physiology , Renal Insufficiency, Chronic/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Cholesterol/metabolism , Cholesterol, LDL/drug effects , Coronary Artery Disease/pathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cresols/pharmacology , Diet, High-Fat , Fecal Microbiota Transplantation , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/microbiology , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Mice , Pinocytosis/drug effects , Renal Insufficiency, Chronic/microbiology , Triglycerides/metabolism
18.
Aquat Toxicol ; 233: 105790, 2021 Apr.
Article En | MEDLINE | ID: mdl-33662879

The potential exposure of titanate nanotubes (TNTs) to wildlife and humans may occur as a result of increased use and application as functional nanomaterials. However, there is a dearth of knowledge regarding the pathways of uptake and excretion of TNTs and their toxicity in cells. In this study, three strains of the Tetrahymena genus of free-living ciliates, including a wild type strain (SB210) and two mutant strains (SB255: mucocyst-deficient; NP1: temperature-sensitive "mouthless''), were used to study the pathways of uptake and excretion and evaluate the cytotoxicity of TNTs. The three Tetrahymena strains were separately exposed to 0, 0.01, 0.1, 1 or 10 mg/L of TNTs, and cells were collected at different time points for quantification of intracellular TNTs (e.g., 5, 10, 20, 40, 60, 90 and 120 min) and evaluation of cytotoxicity (12 and 24 h). TNT contents in NP1 and SB255 were greater or comparable to the contents in SB210 while exposure to 10 mg/L TNTs in 120 min. Furthermore, exposure to 10 mg/L TNTs for 24 h caused greater decreases in cell density of NP1 (38.2 %) and SB255 (36.8 %) compared with SB210 (26.5 %) and upregulated the expression of caspase 15 in SB210. Taken together, our results suggested that TNT uptake by pinocytosis and excretion by exocytosis in Tetrahymena, and the exposure could cause cytotoxicity which can offer novel insights into the accumulation kinetics of nanotubes and even nanomaterials in single cell.


Nanotubes/toxicity , Organisms, Genetically Modified/drug effects , Tetrahymena/drug effects , Titanium/toxicity , Water Pollutants, Chemical/toxicity , Biological Transport , Coloring Agents , Dose-Response Relationship, Drug , Exocytosis/drug effects , Humans , Kinetics , Organisms, Genetically Modified/metabolism , Pinocytosis/drug effects , Tetrahymena/genetics , Tetrahymena/metabolism , Titanium/metabolism , Water Pollutants, Chemical/metabolism
19.
Angew Chem Int Ed Engl ; 60(21): 11928-11936, 2021 05 17.
Article En | MEDLINE | ID: mdl-33629482

Macropinocytosis is a ubiquitous cellular uptake mechanism of peptide-based intracellular delivery. This entry pathway shows promise as a route for the intracellular uptake of biomacromolecules and nanoparticles. In this work, we obtained the 8-residue analogue P4A bearing higher macropinocytosis induction ability. P4A contains vital cysteine residues in its sequence, which immediately reacts with cystine in culture medium to convert into its oxidized forms, including the intramolecularly oxidized form (oxP4A) as the dominant and active species. The conjugate of oxP4A and the membrane lytic peptide LK15 delivered bioactive proteins into cells; notably, this peptide delivered functional proteins fused with a negatively charged protein tag at a significantly reduced amount (up to nanomolar range) without compromising the delivery efficiency and the cellular activities of delivered proteins.


Peptides/metabolism , Pinocytosis/drug effects , Protein Transport/drug effects , Amino Acid Sequence , Cysteine/chemistry , Cysteine/metabolism , Disulfides/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Integrases/metabolism , Peptides/chemistry
20.
Cancer Res ; 81(8): 2086-2100, 2021 04 15.
Article En | MEDLINE | ID: mdl-33593821

Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the "Repurposing" library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2-/- mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2-/- MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2-/- MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2-/- MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. SIGNIFICANCE: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.


Diacylglycerol Kinase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lymphangioleiomyomatosis/drug therapy , Pinocytosis/drug effects , Ritanserin/pharmacology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis/drug therapy , Angiolipoma/genetics , Animals , Autophagy/drug effects , Cell Proliferation , Chloroquine/pharmacology , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Down-Regulation , Drug Synergism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Kidney Neoplasms/genetics , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/etiology , Lymphangioleiomyomatosis/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Nutrients/metabolism , Phosphatidic Acids/metabolism , Phospholipids/metabolism , Pinocytosis/physiology , Tuberous Sclerosis/complications
...