Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38370628

ABSTRACT

DNA-PAINT combined with total Internal Reflection Fluorescence (TIRF) microscopy enables the highest localization precisions, down to single nanometers in thin biological samples, due to TIRF's unique method for optical sectioning and attaining high contrast. However, most cellular targets elude the accessible TIRF range close to the cover glass and thus require alternative imaging conditions, affecting resolution and image quality. Here, we address this limitation by applying ultrathin physical cryosectioning in combination with DNA-PAINT. With "tomographic & kinetically-enhanced" DNA-PAINT (tokPAINT), we demonstrate the imaging of nuclear proteins with sub-3 nanometer localization precision, advancing the quantitative study of nuclear organization within fixed cells and mouse tissues at the level of single antibodies. We believe that ultrathin sectioning combined with the versatility and multiplexing capabilities of DNA-PAINT will be a powerful addition to the toolbox of quantitative DNA-based super-resolution microscopy in intracellular structural analyses of proteins, RNA and DNA in situ.

2.
Med ; 3(2): 119-136, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35425930

ABSTRACT

Background: Ketogenic diet is a potential means of augmenting cancer therapy. Here, we explore ketone body metabolism and its interplay with chemotherapy in pancreatic cancer. Methods: Metabolism and therapeutic responses of murine pancreatic cancer were studied using KPC primary tumors and tumor chunk allografts. Mice on standard high-carbohydrate diet or ketogenic diet were treated with cytotoxic chemotherapy (nab-paclitaxel, gemcitabine, cisplatin). Metabolic activity was monitored with metabolomics and isotope tracing, including 2H- and 13C-tracers, liquid chromatography-mass spectrometry, and imaging mass spectrometry. Findings: Ketone bodies are unidirectionally oxidized to make NADH. This stands in contrast to the carbohydrate-derived carboxylic acids lactate and pyruvate, which rapidly interconvert, buffering NADH/NAD. In murine pancreatic tumors, ketogenic diet decreases glucose's concentration and tricarboxylic acid cycle contribution, enhances 3-hydroxybutyrate's concentration and tricarboxylic acid contribution, and modestly elevates NADH, but does not impact tumor growth. In contrast, the combination of ketogenic diet and cytotoxic chemotherapy substantially raises tumor NADH and synergistically suppresses tumor growth, tripling the survival benefits of chemotherapy alone. Chemotherapy and ketogenic diet also synergize in immune-deficient mice, although long-term growth suppression was only observed in mice with an intact immune system. Conclusions: Ketogenic diet sensitizes murine pancreatic cancer tumors to cytotoxic chemotherapy. Based on these data, we have initiated a randomized clinical trial of chemotherapy with standard versus ketogenic diet for patients with metastatic pancreatic cancer (NCT04631445).


Subject(s)
Diet, Ketogenic , Pancreatic Neoplasms , Animals , Carbohydrates , Diet, Ketogenic/methods , Humans , Mice , NAD , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Pancreatic Neoplasms
3.
Cell Syst ; 13(2): 158-172.e9, 2022 02 16.
Article in English | MEDLINE | ID: mdl-34706266

ABSTRACT

Pancreatic cancer cells with limited access to free amino acids can grow by scavenging extracellular protein. In a murine model of pancreatic cancer, we performed a genome-wide CRISPR screen for genes required for scavenging-dependent growth. The screen identified key mediators of macropinocytosis, peripheral lysosome positioning, endosome-lysosome fusion, lysosomal protein catabolism, and translational control. The top hit was GCN2, a kinase that suppresses translation initiation upon amino acid depletion. Using isotope tracers, we show that GCN2 is not required for protein scavenging. Instead, GCN2 prevents ribosome stalling but without slowing protein synthesis; cells still use all of the limiting amino acids as they emerge from lysosomes. GCN2 also adapts gene expression to the nutrient-poor environment, reorienting protein synthesis away from ribosomes and toward lysosomal hydrolases, such as cathepsin L. GCN2, cathepsin L, and the other genes identified in the screen are potential therapeutic targets in pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Amino Acids/metabolism , Animals , Cathepsin L/metabolism , Mice , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
Nat Protoc ; 16(6): 2802-2825, 2021 06.
Article in English | MEDLINE | ID: mdl-33953394

ABSTRACT

Several essential components of the electron transport chain, the major producer of ATP in mammalian cells, are encoded in the mitochondrial genome. These 13 proteins are translated within mitochondria by 'mitoribosomes'. Defective mitochondrial translation underlies multiple inborn errors of metabolism and has been implicated in pathologies such as aging, metabolic syndrome and cancer. Here, we provide a detailed ribosome profiling protocol optimized to interrogate mitochondrial translation in mammalian cells (MitoRiboSeq), wherein mitoribosome footprints are generated with micrococcal nuclease and mitoribosomes are separated from cytosolic ribosomes and other RNAs by ultracentrifugation in a single straightforward step. We highlight critical steps during library preparation and provide a step-by-step guide to data analysis accompanied by open-source bioinformatic code. Our method outputs mitoribosome footprints at single-codon resolution. Codons with high footprint densities are sites of mitoribosome stalling. We recently applied this approach to demonstrate that defects in mitochondrial serine catabolism or in mitochondrial tRNA methylation cause stalling of mitoribosomes at specific codons. Our method can be applied to study basic mitochondrial biology or to characterize abnormalities in mitochondrial translation in patients with mitochondrial disorders.


Subject(s)
Gene Expression Profiling , Mitochondrial Ribosomes/metabolism , Protein Biosynthesis , Sequence Analysis/methods , HCT116 Cells , Humans
5.
Science ; 360(6390): 710-711, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29773733
6.
Cancer Discov ; 7(11): 1266-1283, 2017 11.
Article in English | MEDLINE | ID: mdl-28899863

ABSTRACT

Lysosomes serve dual roles in cancer metabolism, executing catabolic programs (i.e., autophagy and macropinocytosis) while promoting mTORC1-dependent anabolism. Antimalarial compounds such as chloroquine or quinacrine have been used as lysosomal inhibitors, but fail to inhibit mTOR signaling. Further, the molecular target of these agents has not been identified. We report a screen of novel dimeric antimalarials that identifies dimeric quinacrines (DQ) as potent anticancer compounds, which concurrently inhibit mTOR and autophagy. Central nitrogen methylation of the DQ linker enhances lysosomal localization and potency. An in situ photoaffinity pulldown identified palmitoyl-protein thioesterase 1 (PPT1) as the molecular target of DQ661. PPT1 inhibition concurrently impairs mTOR and lysosomal catabolism through the rapid accumulation of palmitoylated proteins. DQ661 inhibits the in vivo tumor growth of melanoma, pancreatic cancer, and colorectal cancer mouse models and can be safely combined with chemotherapy. Thus, lysosome-directed PPT1 inhibitors represent a new approach to concurrently targeting mTORC1 and lysosomal catabolism in cancer.Significance: This study identifies chemical features of dimeric compounds that increase their lysosomal specificity, and a new molecular target for these compounds, reclassifying these compounds as targeted therapies. Targeting PPT1 blocks mTOR signaling in a manner distinct from catalytic inhibitors, while concurrently inhibiting autophagy, thereby providing a new strategy for cancer therapy. Cancer Discov; 7(11); 1266-83. ©2017 AACR.See related commentary by Towers and Thorburn, p. 1218This article is highlighted in the In This Issue feature, p. 1201.


Subject(s)
Lysosomes/drug effects , Melanoma/drug therapy , Membrane Proteins/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Thiolester Hydrolases/antagonists & inhibitors , Animals , Antimalarials/administration & dosage , Antineoplastic Agents/administration & dosage , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/administration & dosage , Humans , Lysosomes/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Mice , Molecular Targeted Therapy , Proteolysis/drug effects , Signal Transduction/drug effects , Thiolester Hydrolases/genetics
7.
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
8.
Cancer Res ; 75(3): 544-53, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644265

ABSTRACT

Glucose and amino acids are key nutrients supporting cell growth. Amino acids are imported as monomers, but an alternative route induced by oncogenic KRAS involves uptake of extracellular proteins via macropinocytosis and subsequent lysosomal degradation of these proteins as a source of amino acids. In this study, we examined the metabolism of pancreatic ductal adenocarcinoma (PDAC), a poorly vascularized lethal KRAS-driven malignancy. Metabolomic comparisons of human PDAC and benign adjacent tissue revealed that tumor tissue was low in glucose, upper glycolytic intermediates, creatine phosphate, and the amino acids glutamine and serine, two major metabolic substrates. Surprisingly, PDAC accumulated essential amino acids. Such accumulation could arise from extracellular proteins being degraded through macropinocytosis in quantities necessary to meet glutamine requirements, which in turn produces excess of most other amino acids. Consistent with this hypothesis, active macropinocytosis is observed in primary human PDAC specimens. Moreover, in the presence of physiologic albumin, we found that cultured murine PDAC cells grow indefinitely in media lacking single essential amino acids and replicate once in the absence of free amino acids. Growth under these conditions was characterized by simultaneous glutamine depletion and essential amino acid accumulation. Overall, our findings argue that the scavenging of extracellular proteins is an important mode of nutrient uptake in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/metabolism , Albumins/chemistry , Amino Acids/chemistry , Animals , Carcinoma, Pancreatic Ductal/genetics , Cell Proliferation , Chromatography, Liquid , Glucose/chemistry , Glutamine/chemistry , Humans , Mass Spectrometry , Mice , Pinocytosis , Serine/chemistry , Signal Transduction/genetics , Stem Cells/cytology , Tumor Cells, Cultured
9.
Nature ; 497(7451): 633-7, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23665962

ABSTRACT

Macropinocytosis is a highly conserved endocytic process by which extracellular fluid and its contents are internalized into cells through large, heterogeneous vesicles known as macropinosomes. Oncogenic Ras proteins have been shown to stimulate macropinocytosis but the functional contribution of this uptake mechanism to the transformed phenotype remains unknown. Here we show that Ras-transformed cells use macropinocytosis to transport extracellular protein into the cell. The internalized protein undergoes proteolytic degradation, yielding amino acids including glutamine that can enter central carbon metabolism. Accordingly, the dependence of Ras-transformed cells on free extracellular glutamine for growth can be suppressed by the macropinocytic uptake of protein. Consistent with macropinocytosis representing an important route of nutrient uptake in tumours, its pharmacological inhibition compromises the growth of Ras-transformed pancreatic tumour xenografts. These results identify macropinocytosis as a mechanism by which cancer cells support their unique metabolic needs and point to the possible exploitation of this process in the design of anticancer therapies.


Subject(s)
Amino Acids/metabolism , Cell Transformation, Neoplastic , Oncogene Protein p21(ras)/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pinocytosis , Animals , Biological Transport , Carbon/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Female , Glutamine/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Oncogene Protein p21(ras)/genetics , Pancreatic Neoplasms/genetics , Proteolysis
10.
Langmuir ; 27(9): 5335-9, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21456602

ABSTRACT

Fluoride ions play a critical role in preventing tooth decay. We investigated the microscopic effects of fluoride ions on hydroxyapatite (100) surface dissolution using in situ atomic force microscopy. In the presence of 10 mM NaF, individual surface step retraction velocities decreased by about a factor of 5 as compared to NaF-free conditions. Importantly, elongated hexagonal etch pits, which are characteristic of (100) surface dissolution, were no longer observed when NaF was present. The alteration of pit shape is more distinct at a higher NaF concentration (50 mM) where triangular etch pits evolved during dissolution. Furthermore, in a fluoride concentration typical for tap water (10 µM), we observed roughening of individual step lines, resulting in the formation of scalloped morphologies. Morphological changes to individual steps across a wide range of fluoride concentrations suggest that the cariostatic capabilities of fluoride ions originate from their strong interactions with molecular steps.


Subject(s)
Cariostatic Agents/chemistry , Cariostatic Agents/pharmacology , Durapatite/chemistry , Fluorides/chemistry , Fluorides/pharmacology , Microscopy, Atomic Force , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration
SELECTION OF CITATIONS
SEARCH DETAIL
...