Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Cell Chem Biol ; 31(4): 743-759.e8, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38593807

ABSTRACT

Identification of new druggable protein targets remains the key challenge in the current antimalarial development efforts. Here we used mass-spectrometry-based cellular thermal shift assay (MS-CETSA) to identify potential targets of several antimalarials and drug candidates. We found that falcilysin (FLN) is a common binding partner for several drug candidates such as MK-4815, MMV000848, and MMV665806 but also interacts with quinoline drugs such as chloroquine and mefloquine. Enzymatic assays showed that these compounds can inhibit FLN proteolytic activity. Their interaction with FLN was explored systematically by isothermal titration calorimetry and X-ray crystallography, revealing a shared hydrophobic pocket in the catalytic chamber of the enzyme. Characterization of transgenic cell lines with lowered FLN expression demonstrated statistically significant increases in susceptibility toward MK-4815, MMV000848, and several quinolines. Importantly, the hydrophobic pocket of FLN appears amenable to inhibition and the structures reported here can guide the development of novel drugs against malaria.


Subject(s)
Antimalarials , Malaria , Methylamines , Quinolines , Humans , Antimalarials/chemistry , Malaria/drug therapy , Phenols/therapeutic use , Quinolines/pharmacology , Quinolines/metabolism , Drug Development
2.
Sci Rep ; 14(1): 1878, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38253642

ABSTRACT

Mass spectrometry-coupled cellular thermal shift assay (MS-CETSA), a biophysical principle-based technique that measures the thermal stability of proteins at the proteome level inside the cell, has contributed significantly to the understanding of drug mechanisms of action and the dissection of protein interaction dynamics in different cellular states. One of the barriers to the wide applications of MS-CETSA is that MS-CETSA experiments must be performed on the specific cell lines of interest, which is typically time-consuming and costly in terms of labeling reagents and mass spectrometry time. In this study, we aim to predict CETSA features in various cell lines by introducing a computational framework called CycleDNN based on deep neural network technology. For a given set of n cell lines, CycleDNN comprises n auto-encoders. Each auto-encoder includes an encoder to convert CETSA features from one cell line into latent features in a latent space [Formula: see text]. It also features a decoder that transforms the latent features back into CETSA features for another cell line. In such a way, the proposed CycleDNN creates a cyclic prediction of CETSA features across different cell lines. The prediction loss, cycle-consistency loss, and latent space regularization loss are used to guide the model training. Experimental results on a public CETSA dataset demonstrate the effectiveness of our proposed approach. Furthermore, we confirm the validity of the predicted MS-CETSA data from our proposed CycleDNN through validation in protein-protein interaction prediction.


Subject(s)
Deep Learning , Biophysics , Cell Line , Dissection , Mass Spectrometry
3.
Acta Pharmacol Sin ; 45(2): 391-404, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37803139

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common and deadly cancers in the world. The therapeutic outlook for HCC patients has significantly improved with the advent and development of systematic and targeted therapies such as sorafenib and lenvatinib; however, the rise of drug resistance and the high mortality rate necessitate the continuous discovery of effective targeting agents. To discover novel anti-HCC compounds, we first constructed a deep learning-based chemical representation model to screen more than 6 million compounds in the ZINC15 drug-like library. We successfully identified LGOd1 as a novel anticancer agent with a characteristic levoglucosenone (LGO) scaffold. The mechanistic studies revealed that LGOd1 treatment leads to HCC cell death by interfering with cellular copper homeostasis, which is similar to a recently reported copper-dependent cell death named cuproptosis. While the prototypical cuproptosis is brought on by copper ionophore-induced copper overload, mechanistic studies indicated that LGOd1 does not act as a copper ionophore, but most likely by interacting with the copper chaperone protein CCS, thus LGOd1 represents a potentially new class of compounds with unique cuproptosis-inducing property. In summary, our findings highlight the critical role of bioavailable copper in the regulation of cell death and represent a novel route of cuproptosis induction.


Subject(s)
Carcinoma, Hepatocellular , Deep Learning , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Copper , Liver Neoplasms/drug therapy , Ionophores , Apoptosis
4.
Annu Int Conf IEEE Eng Med Biol Soc ; 2022: 1659-1662, 2022 07.
Article in English | MEDLINE | ID: mdl-36085889

ABSTRACT

The Cellular Thermal Shift Assay (CETSA) is a biophysical assay based on the principle of ligand-induced thermal stabilization of target proteins. This technology has revolutionized cell-based target engagement studies and has been used as guidance for drug design. Although many ap-plications of CETSA data have been explored, the correlations between CETSA data and protein-protein interactions (PPI) have barely been touched. In this study, we conduct the first exploration study applying CETSA data for PPI prediction. We use a machine learning method, Decision Tree, to predict PPI scores using proteins' CETSA features. It shows promising results that the predicted PPI scores closely match the ground-truth PPI scores. Furthermore, for a small number of protein pairs, whose PPI score predictions mismatch the ground truth, we use iterative clustering strategy to gradually reduce the number of these pairs. At the end of iterative clustering, the remaining protein pairs may have some unusual properties and are of scientific value for further biological investigation. Our study has demonstrated that PPI is a brand-new application of CETSA data. At the same time, it also manifests that CETSA data can be used as a new data source for PPI exploration study.


Subject(s)
Biological Assay , Research Design , Biophysics , Cluster Analysis , Protein Domains
5.
Annu Int Conf IEEE Eng Med Biol Soc ; 2022: 1647-1650, 2022 07.
Article in English | MEDLINE | ID: mdl-36085941

ABSTRACT

Cellular Thermal Shift Assay (CETSA) has been widely used in drug discovery, cancer cell biology, immunology, etc. One of the barriers for CETSA applications is that CETSA experiments have to be conducted on various cell lines, which is extremely time-consuming and costly. In this study, we make an effort to explore the translation of CETSA features cross cell lines, i.e., known CETSA feature of a given protein in one cell line, can we automatically predict the CETSA feature of this protein in another cell line, and vice versa? Inspired by pix2pix and CycleGAN, which perform well on image-to-image translation cross various domains in computer vision, we propose a novel deep neural network model called CycleDNN for CETSA feature translation cross cell lines. Given cell lines A and B, the proposed CycleDNN consists of two auto-encoders, the first one encodes the CETSA feature from cell line A into Z in the latent space [Formula: see text], then decodes Z into the CETSA feature in cell line B., Similarly, the second one translates the CETSA feature from cell line B to cell line A through the latent space [Formula: see text]. In such a way, the two auto-encoders form a cyclic feature translation between cell lines. The reconstructed loss, cycle-consistency loss, and latent vector regularization loss are used to guide the training of the model. The experimental results on a public CETSA dataset demonstrate the effectiveness of the proposed approach.


Subject(s)
Drug Discovery , Neural Networks, Computer , Cell Line , Drug Discovery/methods , Proteins , Research Design
6.
Cell Chem Biol ; 29(4): 572-585.e8, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34265272

ABSTRACT

The optimal use of many cancer drugs is hampered by a lack of detailed understanding of their mechanism of action (MoA). Here, we apply a high-resolution implementation of the proteome-wide cellular thermal shift assay (CETSA) to follow protein interaction changes induced by the antimetabolite 5-fluorouracil (5-FU) and related nucleosides. We confirm anticipated effects on the known main target, thymidylate synthase (TYMS), and enzymes in pyrimidine metabolism and DNA damage pathways. However, most interaction changes we see are for proteins previously not associated with the MoA of 5-FU, including wide-ranging effects on RNA-modification and -processing pathways. Attenuated responses of specific proteins in a resistant cell model identify key components of the 5-FU MoA, where intriguingly the abrogation of TYMS inhibition is not required for cell proliferation.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Fluorouracil/pharmacology , Proteome , Proteomics , RNA
7.
Med Res Rev ; 41(6): 2893-2926, 2021 11.
Article in English | MEDLINE | ID: mdl-33533067

ABSTRACT

Small-molecule drugs modulate biological processes and disease states through engagement of target proteins in cells. Assessing drug-target engagement on a proteome-wide scale is of utmost importance in better understanding the molecular mechanisms of action of observed beneficial and adverse effects, as well as in developing next generation tool compounds and drugs with better efficacies and specificities. However, systematic assessment of drug-target engagement has been an arduous task. With the continuous development of mass spectrometry-based proteomics instruments and techniques, various chemical proteomics approaches for drug target deconvolution (i.e., the identification of molecular target for drugs) have emerged. Among these, the label-free target deconvolution approaches that do not involve the chemical modification of compounds of interest, have gained increased attention in the community. Here we provide an overview of the basic principles and recent biological applications of the most important label-free methods including the cellular thermal shift assay, pulse proteolysis, chemical denaturant and protein precipitation, stability of proteins from rates of oxidation, drug affinity responsive target stability, limited proteolysis, and solvent-induced protein precipitation. The state-of-the-art technical implications and future outlook for the label-free approaches are also discussed.


Subject(s)
Proteome , Proteomics , Drug Delivery Systems , Humans , Oxidation-Reduction , Proteome/metabolism , Proteomics/methods , Solvents
8.
Nat Protoc ; 15(6): 1881-1921, 2020 06.
Article in English | MEDLINE | ID: mdl-32341577

ABSTRACT

Despite decades of research, little is known about the cellular targets and the mode of action of the vast majority of antimalarial drugs. We recently demonstrated that the cellular thermal shift assay (CETSA) protocol in its two variants: the melt curve and the isothermal dose-response, represents a comprehensive strategy for the identification of antimalarial drug targets. CETSA enables proteome-wide target screening for unmodified antimalarial compounds with undetermined mechanisms of action, providing quantitative evidence about direct drug-protein interactions. The experimental workflow involves treatment of P. falciparum-infected erythrocytes with a compound of interest, heat exposure to denature proteins, soluble protein isolation, enzymatic digestion, peptide labeling with tandem mass tags, offline fractionation, and liquid chromatography-tandem mass spectrometry analysis. Methodological optimizations necessary for the analysis of this intracellular parasite are discussed, including enrichment of parasitized cells and hemoglobin depletion strategies to overcome high hemoglobin abundance in the host red blood cells. We outline an effective data processing workflow using the mineCETSA R package, which enables prioritization of drug-target candidates for follow-up studies. The entire protocol can be completed within 2 weeks.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Protozoan Proteins/metabolism , Drug Discovery/methods , Erythrocytes/parasitology , Humans , Malaria, Falciparum/metabolism , Molecular Targeted Therapy/methods , Parasitic Sensitivity Tests/methods , Plasmodium falciparum/metabolism , Proteome/metabolism
9.
Curr Opin Chem Biol ; 54: 54-62, 2020 02.
Article in English | MEDLINE | ID: mdl-31838273

ABSTRACT

The Cellular Thermal Shift Assay (CETSA) has recently emerged as a promising method to directly monitor functional modulations of protein interaction states in intact cells and tissue samples. Recent data support that the mass spectrometry-coupled proteome-wide implementation of CETSA (MS-CETSA) generates stringent information on a wide range of different interaction classes and is uniquely well suited to study the modulation of protein interaction states in cellular processes and during drug action. To expand the mechanistic insight of CETSA shifts, and to complement information from CETSA experiments, we outline how the integration of MS-CETSA with other proteomics techniques can provide a new platform for detailed, comprehensive, and interactive studies of the functional modulations of proteomes in situ.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Proteomics/methods , Humans , Metabolomics/methods , Protein Interaction Maps , Tandem Mass Spectrometry
10.
Annu Rev Biochem ; 88: 383-408, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30939043

ABSTRACT

The cellular thermal shift assay (CETSA) is a biophysical technique allowing direct studies of ligand binding to proteins in cells and tissues. The proteome-wide implementation of CETSA with mass spectrometry detection (MS-CETSA) has now been successfully applied to discover targets for orphan clinical drugs and hits from phenotypic screens, to identify off-targets, and to explain poly-pharmacology and drug toxicity. Highly sensitive multidimensional MS-CETSA implementations can now also access binding of physiological ligands to proteins, such as metabolites, nucleic acids, and other proteins. MS-CETSA can thereby provide comprehensive information on modulations of protein interaction states in cellular processes, including downstream effects of drugs and transitions between different physiological cell states. Such horizontal information on ligandmodulation in cells is largely orthogonal to vertical information on the levels of different proteins and therefore opens novel opportunities to understand operational aspects of cellular proteomes.


Subject(s)
Drug Development/methods , Proteome/metabolism , Electrophoretic Mobility Shift Assay , Humans , Ligands , Mass Spectrometry , Protein Binding , Proteome/chemistry , Proteomics
11.
Redox Biol ; 24: 101168, 2019 06.
Article in English | MEDLINE | ID: mdl-30925293

ABSTRACT

Reactive oxygen species (ROS) induce different cellular stress responses but can also mediate cellular signaling. Augmented levels of ROS are associated with aging, cancer as well as various metabolic and neurological disorders. ROS can also affect the efficacy and adverse effects of drugs. Although proteins are key mediators of most ROS effects, direct studies of ROS-modulated-protein function in the cellular context are very challenging. Therefore the understanding of specific roles of different proteins in cellular ROS responses is still relatively rudimentary. In the present work we show that Mass Spectrometry-Cellular Thermal Shift Assay (MS-CETSA) can directly monitor ROS and redox modulations of protein structure at the proteome level. By altering ROS levels in cultured human hepatocellular carcinoma cell lysates and intact cells, we detected CETSA responses in many proteins known to be redox sensitive, and also revealed novel candidate ROS sensitive proteins. Studies in intact cells treated with hydrogen peroxide and sulfasalazine, a ROS modulating drug, identified not only proteins that are directly modified, but also proteins reporting on downstream cellular effects. Comprehensive changes are seen on rate-limiting proteins regulating key cellular processes, including known redox control systems, protein degradation, epigenetic control and protein translational processes. Interestingly, concerted shifts on ATP-binding proteins revealed redox-induced modulation of ATP levels, which likely control many cellular processes. Collectively, these studies establish CETSA as a novel method for cellular studies of redox modulations of proteins, which implicated in a wide range of processes and for the discovery of CETSA-based biomarkers reporting on the efficacy as well as adverse effects of drugs.


Subject(s)
Oxidation-Reduction , Proteins/chemistry , Proteins/metabolism , Proteomics , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Biomarkers , Cell Line, Tumor , Chromatography, Liquid , Glutathione/metabolism , Humans , Hydrogen Peroxide/chemistry , Mass Spectrometry , Proteomics/methods , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Workflow
12.
Sci Transl Med ; 11(473)2019 01 02.
Article in English | MEDLINE | ID: mdl-30602534

ABSTRACT

Mechanisms of action (MoAs) have been elusive for most antimalarial drugs in clinical use. Decreasing responsiveness to antimalarial treatments stresses the need for a better resolved understanding of their MoAs and associated resistance mechanisms. In the present work, we implemented the cellular thermal shift assay coupled with mass spectrometry (MS-CETSA) for drug target identification in Plasmodium falciparum, the main causative agent of human malaria. We validated the efficacy of this approach for pyrimethamine, a folic acid antagonist, and E64d, a broad-spectrum cysteine proteinase inhibitor. Subsequently, we applied MS-CETSA to quinine and mefloquine, two important antimalarial drugs with poorly characterized MoAs. Combining studies in the P. falciparum parasite lysate and intact infected red blood cells, we found P. falciparum purine nucleoside phosphorylase (PfPNP) as a common binding target for these two quinoline drugs. Biophysical and structural studies with a recombinant protein further established that both compounds bind within the enzyme's active site. Quinine binds to PfPNP at low nanomolar affinity, suggesting a substantial contribution to its therapeutic effect. Overall, we demonstrated that implementation of MS-CETSA for P. falciparum constitutes a promising strategy to elucidate the MoAs of existing and candidate antimalarial drugs.


Subject(s)
Biological Assay/methods , Molecular Targeted Therapy , Purine-Nucleoside Phosphorylase/metabolism , Quinine/pharmacology , Temperature , Mefloquine/pharmacology , Models, Molecular , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Proteome/metabolism , Reproducibility of Results
13.
Contemp Clin Dent ; 10(3): 577-581, 2019.
Article in English | MEDLINE | ID: mdl-32308341

ABSTRACT

Patients afflicted with congenital cleft lip defect experience disfigured facial esthetics and lack of oral competency that could reduce the speech intelligibility. Rehabilitation of such cosmetic deformities condition often encountered with dental abnormalities becomes the challenge. When surgical treatment is not desired, prosthetic reconstruction turns out to be an economical option. This clinical treatment describes the fabrication of a maxillary lip prosthesis supported by a cast partial denture retained with magnetic attachment in a cleft lip patient. The use of intraoral magnets placed labially on the cast partial denture and on the counterpart of the prosthesis improved the retention. Moreover, the biomedical-grade silicone prosthesis used to blend closely with the patient's skin shade fulfilled the esthetic needs.

14.
PLoS One ; 13(12): e0208273, 2018.
Article in English | MEDLINE | ID: mdl-30521565

ABSTRACT

Metabolite-protein interactions define the output of metabolic pathways and regulate many cellular processes. Although diseases are often characterized by distortions in metabolic processes, efficient means to discover and study such interactions directly in cells have been lacking. A stringent implementation of proteome-wide Cellular Thermal Shift Assay (CETSA) was developed and applied to key cellular nucleotides, where previously experimentally confirmed protein-nucleotide interactions were well recaptured. Many predicted, but never experimentally confirmed, as well as novel protein-nucleotide interactions were discovered. Interactions included a range of different protein families where nucleotides serve as substrates, products, co-factors or regulators. In cells exposed to thymidine, a limiting precursor for DNA synthesis, both dose- and time-dependence of the intracellular binding events for sequentially generated thymidine metabolites were revealed. Interactions included known cancer targets in deoxyribonucleotide metabolism as well as novel interacting proteins. This stringent CETSA based strategy will be applicable for a wide range of metabolites and will therefore greatly facilitate the discovery and studies of interactions and specificities of the many metabolites in human cells that remain uncharacterized.


Subject(s)
Nucleotides/metabolism , Proteins/metabolism , Proteome/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nucleotides/genetics , Protein Binding , Proteins/genetics , Proteome/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
15.
Indian J Dent Res ; 29(3): 352-357, 2018.
Article in English | MEDLINE | ID: mdl-29900921

ABSTRACT

PURPOSE: This study investigated and compared the surface roughness achieved by glazing porcelain samples in a conventional and a microwave oven. MATERIALS AND METHODS: Two commercial brands of metal ceramics were used, VITA VMK MASTER and IPS CLASSIC. Sixty samples were fabricated, 30 for each type of ceramic. The samples were sintered in the conventional oven and hand-polished to remove any irregularities. Samples (n = 10) from each type of ceramic were further divided into three groups as follows: hand-polished (Group A), conventional oven glazed (Group B), and microwave glazed (Group C). Each specimen was evaluated for surface roughness by atomic force microscope. Data were statistically analyzed using two-way analysis of variance (ANOVA) and Tukey's post hoc test (a = 0.05). RESULTS: Two-way ANOVA indicated a highly significant difference in surface roughness based on the type of glazing (P < 0.001), there was a significant difference based on the metal ceramics (P = 0.002). There was also a significant interaction between the type of glazing and metal ceramics (P = 0.009). The images obtained from the atomic force microscope corroborated the measured values. CONCLUSIONS: All the results indicate that microwave glazing can be a feasible option for glazing porcelain specimens. It was concluded that surface topography is influenced by surface treatment and microwave glazed ceramic is superior to conventional oven glazed ceramic and hand-polishing showed greater surface roughness when compared to glazing. IPS CLASSIC ceramic showed relatively smooth surface when compared to VITA VMK MASTER irrespective of the surface treatment.


Subject(s)
Dental Polishing/methods , Dental Porcelain/chemistry , Microscopy, Atomic Force , Microwaves , Materials Testing , Surface Properties
16.
Cell ; 173(6): 1481-1494.e13, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29706543

ABSTRACT

Global profiling of protein expression through the cell cycle has revealed subsets of periodically expressed proteins. However, expression levels alone only give a partial view of the biochemical processes determining cellular events. Using a proteome-wide implementation of the cellular thermal shift assay (CETSA) to study specific cell-cycle phases, we uncover changes of interaction states for more than 750 proteins during the cell cycle. Notably, many protein complexes are modulated in specific cell-cycle phases, reflecting their roles in processes such as DNA replication, chromatin remodeling, transcription, translation, and disintegration of the nuclear envelope. Surprisingly, only small differences in the interaction states were seen between the G1 and the G2 phase, suggesting similar hardwiring of biochemical processes in these two phases. The present work reveals novel molecular details of the cell cycle and establishes proteome-wide CETSA as a new strategy to study modulation of protein-interaction states in intact cells.


Subject(s)
Cell Cycle , Protein Interaction Mapping , Cell Division , Chromatin/chemistry , Cluster Analysis , DNA Replication , G1 Phase , G2 Phase , Humans , K562 Cells , Nuclear Envelope , Proteome , Proteomics/methods
17.
Contemp Clin Dent ; 9(1): 120-122, 2018.
Article in English | MEDLINE | ID: mdl-29599597

ABSTRACT

The restoration of normal function and esthetics is often challenging in the prosthetic rehabilitation of patients with hemimandibulectomy defects due to unstable occlusion and mandibular deviation. The extensive period of time for completion of healing of the reconstructed mandible through reconstructive plastic surgery and/or implant-assisted prosthesis may compromise the masticatory function by causing delay in the fabrication of definitive prosthesis. This case report describes a novel technique for the construction of customized attachment-retained mandibular guiding flange prosthesis for immediate rectification of the frontal plane rotation occurring after hemimandibulectomy.

18.
Science ; 359(6380): 1170-1177, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29439025

ABSTRACT

Proteins differentially interact with each other across cellular states and conditions, but an efficient proteome-wide strategy to monitor them is lacking. We report the application of thermal proximity coaggregation (TPCA) for high-throughput intracellular monitoring of protein complex dynamics. Significant TPCA signatures observed among well-validated protein-protein interactions correlate positively with interaction stoichiometry and are statistically observable in more than 350 annotated human protein complexes. Using TPCA, we identified many complexes without detectable differential protein expression, including chromatin-associated complexes, modulated in S phase of the cell cycle. Comparison of six cell lines by TPCA revealed cell-specific interactions even in fundamental cellular processes. TPCA constitutes an approach for system-wide studies of protein complexes in nonengineered cells and tissues and might be used to identify protein complexes that are modulated in diseases.


Subject(s)
Multiprotein Complexes/metabolism , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Cell Line , Cells , Chromatin/metabolism , Hot Temperature , Humans , Protein Array Analysis , Protein Biosynthesis , Protein Folding , Proteome
19.
PLoS One ; 13(1): e0190063, 2018.
Article in English | MEDLINE | ID: mdl-29293541

ABSTRACT

Influenza and asthma are two of the major public health concerns in the world today. During the 2009 influenza pandemic asthma was found to be the commonest comorbid illness of patients admitted to hospital. Unexpectedly, it was also observed that asthmatic patients admitted to hospital with influenza infection were less likely to die or require admission to intensive care compared with non-asthmatics. Using an in vivo model of asthma and influenza infection we demonstrate that prior exposure to Blomia tropicalis extract (BTE) leads to an altered immune response to influenza infection, comprised of less severe weight loss and faster recovery following infection. This protection was associated with significant increases in T cell numbers in the lungs of BTE sensitised and infected mice, as well as increased IFN-γ production from these cells. In addition, elevated numbers of CD11b+ dendritic cells (DCs) were found in the lung draining lymph nodes following infection of BTE sensitised mice compared to infected PBS treated mice. These CD11b+ DCs appeared to be better at priming CD8 specific T cells both in vivo and ex vivo, a function not normally attributed to CD11b+ DCs. We propose that this alteration in cross-presentation and more efficient T cell priming seen in BTE sensitised mice, led to the earlier increase in T cells in the lungs and subsequently faster clearance of the virus and reduced influenza induced pathology. We believe this data provides a novel mechanism that explains why asthmatic patients may present with less severe disease when infected with influenza.


Subject(s)
Allergens/administration & dosage , Asthma/immunology , Dendritic Cells/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Animals , Asthma/complications , Bronchoalveolar Lavage Fluid , Dose-Response Relationship, Drug , Flow Cytometry , Inhalation Exposure , Interferon-gamma/biosynthesis , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology , Plant Extracts/pharmacology , Viral Load
20.
Nat Commun ; 8(1): 2200, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259156

ABSTRACT

Achieving robust cancer-specific lethality is the ultimate clinical goal. Here, we identify a compound with dual-inhibitory properties, named a131, that selectively kills cancer cells, while protecting normal cells. Through an unbiased CETSA screen, we identify the PIP4K lipid kinases as the target of a131. Ablation of the PIP4Ks generates a phenocopy of the pharmacological effects of PIP4K inhibition by a131. Notably, PIP4Ks inhibition by a131 causes reversible growth arrest in normal cells by transcriptionally upregulating PIK3IP1, a suppressor of the PI3K/Akt/mTOR pathway. Strikingly, Ras activation overrides a131-induced PIK3IP1 upregulation and activates the PI3K/Akt/mTOR pathway. Consequently, Ras-transformed cells override a131-induced growth arrest and enter mitosis where a131's ability to de-cluster supernumerary centrosomes in cancer cells eliminates Ras-activated cells through mitotic catastrophe. Our discovery of drugs with a dual-inhibitory mechanism provides a unique pharmacological strategy against cancer and evidence of cross-activation between the Ras/Raf/MEK/ERK and PI3K/AKT/mTOR pathways via a Ras˧PIK3IP1˧PI3K signaling network.


Subject(s)
Acrylonitrile/analogs & derivatives , Indoles/pharmacology , Isoquinolines/pharmacology , Membrane Proteins/metabolism , Mitosis/drug effects , Neoplasms/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Acrylonitrile/pharmacology , Acrylonitrile/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Indoles/therapeutic use , Intracellular Signaling Peptides and Proteins , Isoquinolines/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...