Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
2.
ACS Pharmacol Transl Sci ; 5(11): 1156-1168, 2022 Nov 11.
Article in English | MEDLINE | ID: mdl-36407952

ABSTRACT

Bruton's tyrosine kinase (BTK) is a member of the TEC-family kinases and crucial for the proliferation and differentiation of B-cells. We evaluated the therapeutic potential of a covalent inhibitor (JS25) with nanomolar potency against BTK and with a more desirable selectivity and inhibitory profile compared to the FDA-approved BTK inhibitors ibrutinib and acalabrutinib. Structural prediction of the BTK/JS25 complex revealed sequestration of Tyr551 that leads to BTK's inactivation. JS25 also inhibited the proliferation of myeloid and lymphoid B-cell cancer cell lines. Its therapeutic potential was further tested against ibrutinib in preclinical models of B-cell cancers. JS25 treatment induced a more pronounced cell death in a murine xenograft model of Burkitt's lymphoma, causing a 30-40% reduction of the subcutaneous tumor and an overall reduction in the percentage of metastasis and secondary tumor formation. In a patient model of diffuse large B-cell lymphoma, the drug response of JS25 was higher than that of ibrutinib, leading to a 64% "on-target" efficacy. Finally, in zebrafish patient-derived xenografts of chronic lymphocytic leukemia, JS25 was faster and more effective in decreasing tumor burden, producing superior therapeutic effects compared to ibrutinib. We expect JS25 to become therapeutically relevant as a BTK inhibitor and to find applications in the treatment of hematological cancers and other pathologies with unmet clinical treatment.

3.
Blood Cancer Discov ; 3(6): 502-515, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36125297

ABSTRACT

Drug testing in patient biopsy-derived cells can identify potent treatments for patients suffering from relapsed or refractory hematologic cancers. Here we investigate the use of weakly supervised deep learning on cell morphologies (DML) to complement diagnostic marker-based identification of malignant and nonmalignant cells in drug testing. Across 390 biopsies from 289 patients with diverse blood cancers, DML-based drug responses show improved reproducibility and clustering of drugs with the same mode of action. DML does so by adapting to batch effects and by autonomously recognizing disease-associated cell morphologies. In a post hoc analysis of 66 patients, DML-recommended treatments led to improved progression-free survival compared with marker-based recommendations and physician's choice-based treatments. Treatments recommended by both immunofluorescence and DML doubled the fraction of patients achieving exceptional clinical responses. Thus, DML-enhanced ex vivo drug screening is a promising tool in the identification of effective personalized treatments. SIGNIFICANCE: We have recently demonstrated that image-based drug screening in patient samples identifies effective treatment options for patients with advanced blood cancers. Here we show that using deep learning to identify malignant and nonmalignant cells by morphology improves such screens. The presented workflow is robust, automatable, and compatible with clinical routine. This article is highlighted in the In This Issue feature, p. 476.


Subject(s)
Hematologic Neoplasms , Precision Medicine , Humans , Reproducibility of Results
4.
Cancer Discov ; 12(2): 372-387, 2022 02.
Article in English | MEDLINE | ID: mdl-34635570

ABSTRACT

Personalized medicine aims to match the right drug with the right patient by using specific features of the individual patient's tumor. However, current strategies of personalized therapy matching provide treatment opportunities for less than 10% of patients with cancer. A promising method may be drug profiling of patient biopsy specimens with single-cell resolution to directly quantify drug effects. We prospectively tested an image-based single-cell functional precision medicine (scFPM) approach to guide treatments in 143 patients with advanced aggressive hematologic cancers. Fifty-six patients (39%) were treated according to scFPM results. At a median follow-up of 23.9 months, 30 patients (54%) demonstrated a clinical benefit of more than 1.3-fold enhanced progression-free survival compared with their previous therapy. Twelve patients (40% of responders) experienced exceptional responses lasting three times longer than expected for their respective disease. We conclude that therapy matching by scFPM is clinically feasible and effective in advanced aggressive hematologic cancers. SIGNIFICANCE: This is the first precision medicine trial using a functional assay to instruct n-of-one therapies in oncology. It illustrates that for patients lacking standard therapies, high-content assay-based scFPM can have a significant value in clinical therapy guidance based on functional dependencies of each patient's cancer.See related commentary by Letai, p. 290.This article is highlighted in the In This Issue feature, p. 275.


Subject(s)
Hematologic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Austria , Cohort Studies , Female , Hematologic Neoplasms/mortality , Humans , Male , Middle Aged , Molecular Targeted Therapy , Precision Medicine , Progression-Free Survival , Young Adult
5.
Curr Opin Chem Biol ; 56: 72-78, 2020 06.
Article in English | MEDLINE | ID: mdl-32086157

ABSTRACT

Anticancer drug discovery and development using conventional cell line and animal models has traditionally had a low overall success rate. Despite yielding game-changing new therapeutics, 10-20 new molecules have to be brought to the clinic to obtain one new approval, making this approach costly and inefficient. The use of in vitro experimental models based on primary human tumour tissues has the potential to provide a representation of human cancer biology that is closer to an actual patient and to 'bridge the translational gap' between preclinical and clinical research. Here, we review recent advances in the use of human tumour samples for preclinical research through organoid development or as primary patient materials. While challenges still remain regarding analysis, validation and scalability, evidence is mounting for the applicability of both models as preclinical research tools.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/diagnostic imaging , Neoplasms/therapy , Animals , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Machine Learning , Models, Biological , Optical Imaging , Organoids/metabolism , Single-Cell Analysis
6.
Haematologica ; 104(10): 1935-1949, 2019 10.
Article in English | MEDLINE | ID: mdl-31048353

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a myeloid neoplasm characterized by dysplasia, abnormal production and accumulation of monocytic cells and an elevated risk of transforming into acute leukemia. Over the past two decades, our knowledge about the pathogenesis and molecular mechanisms in CMML has increased substantially. In parallel, better diagnostic criteria and therapeutic strategies have been developed. However, many questions remain regarding prognostication and optimal therapy. In addition, there is a need to define potential pre-phases of CMML and special CMML variants, and to separate these entities from each other and from conditions mimicking CMML. To address these unmet needs, an international consensus group met in a Working Conference in August 2018 and discussed open questions and issues around CMML, its variants, and pre-CMML conditions. The outcomes of this meeting are summarized herein and include diag nostic criteria and a proposed classification of pre-CMML conditions as well as refined minimal diagnostic criteria for classical CMML and special CMML variants, including oligomonocytic CMML and CMML associated with systemic mastocytosis. Moreover, we propose diagnostic standards and tools to distinguish between 'normal', pre-CMML and CMML entities. These criteria and standards should facilitate diagnostic and prognostic evaluations in daily practice and clinical studies in applied hematology.


Subject(s)
Leukemia, Myelomonocytic, Chronic/diagnosis , Precancerous Conditions/diagnosis , Aged , Congresses as Topic , Female , Humans , Male , Middle Aged , Practice Guidelines as Topic
7.
Nat Chem Biol ; 15(3): 232-240, 2019 03.
Article in English | MEDLINE | ID: mdl-30692684

ABSTRACT

The Bruton tyrosine kinase (BTK) inhibitor ibrutinib has substantially improved therapeutic options for chronic lymphocytic leukemia (CLL). Although ibrutinib is not curative, it has a profound effect on CLL cells and may create new pharmacologically exploitable vulnerabilities. To identify such vulnerabilities, we developed a systematic approach that combines epigenome profiling (charting the gene-regulatory basis of cell state) with single-cell chemosensitivity profiling (quantifying cell-type-specific drug response) and bioinformatic data integration. By applying our method to a cohort of matched patient samples collected before and during ibrutinib therapy, we identified characteristic ibrutinib-induced changes that provide a starting point for the rational design of ibrutinib combination therapies. Specifically, we observed and validated preferential sensitivity to proteasome, PLK1, and mTOR inhibitors during ibrutinib treatment. More generally, our study establishes a broadly applicable method for investigating treatment-specific vulnerabilities by integrating the complementary perspectives of epigenetic cell states and phenotypic drug responses in primary patient samples.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Chromatin/physiology , Drug Combinations , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Piperidines , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Single-Cell Analysis/methods , TOR Serine-Threonine Kinases/metabolism , Polo-Like Kinase 1
8.
Lab Invest ; 99(5): 648-658, 2019 05.
Article in English | MEDLINE | ID: mdl-30679758

ABSTRACT

Fibroblast-like synoviocytes (FLS) are major contributors to joint inflammation in rheumatoid arthritis (RA). Forkhead box O 3 (FOXO3) perturbations in immune cells are increasingly linked to RA pathogenesis. Here, we show that FOXO3 is distinctly inactivated/phosphorylated in the FLS of rheumatoid synovitis. In vitro, stimulation of FLS with tumor necrosis factor-alpha α (TNFα) induced a rapid and sustained inactivation of FOXO3. mRNA profiling revealed that the inactivation of FOXO3 is important for the sustained pro-inflammatory interferon response to TNFα (CXCL9, CXCL10, CXCL11, and TNFSF18). Mechanistically, our studies demonstrate that the inactivation of FOXO3 results from TNF-induced downregulation of phosphoinositide-3-kinase-interacting protein 1 (PIK3IP1). Thus, we identified FOXO3 and its modulator PIK3IP1 as a critical regulatory circuit for the inflammatory response of the resident mesenchymal cells to TNFα and contribute insight into how the synovial tissue brings about chronic inflammation that is driven by TNFα.


Subject(s)
Fibroblasts/drug effects , Forkhead Box Protein O3/genetics , Inflammation/genetics , Synoviocytes/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Forkhead Box Protein O3/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Synoviocytes/cytology , Synoviocytes/metabolism
9.
Science ; 362(6419): 1171-1177, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30442766

ABSTRACT

In genetic screens aimed at understanding drug resistance mechanisms in chronic myeloid leukemia cells, inactivation of the cullin 3 adapter protein-encoding leucine zipper-like transcription regulator 1 (LZTR1) gene led to enhanced mitogen-activated protein kinase (MAPK) pathway activity and reduced sensitivity to tyrosine kinase inhibitors. Knockdown of the Drosophila LZTR1 ortholog CG3711 resulted in a Ras-dependent gain-of-function phenotype. Endogenous human LZTR1 associates with the main RAS isoforms. Inactivation of LZTR1 led to decreased ubiquitination and enhanced plasma membrane localization of endogenous KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). We propose that LZTR1 acts as a conserved regulator of RAS ubiquitination and MAPK pathway activation. Because LZTR1 disease mutations failed to revert loss-of-function phenotypes, our findings provide a molecular rationale for LZTR1 involvement in a variety of inherited and acquired human disorders.


Subject(s)
Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/physiology , Ubiquitination , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drosophila melanogaster , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gain of Function Mutation , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/epidemiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Loss of Function Mutation , MAP Kinase Signaling System/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Signal Transduction , Transcription Factors/genetics , Ubiquitination/genetics
10.
Lancet Haematol ; 4(12): e595-e606, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29153976

ABSTRACT

BACKGROUND: Patients with refractory or relapsed haematological malignancies have few treatment options and short survival times. Identification of effective therapies with genomic-based precision medicine is hampered by intratumour heterogeneity and incomplete understanding of the contribution of various mutations within specific cancer phenotypes. Ex-vivo drug-response profiling in patient biopsies might aid effective treatment identification; however, proof of its clinical utility is limited. METHODS: We investigated the feasibility and clinical impact of multiparametric, single-cell, drug-response profiling in patient biopsies by immunofluorescence, automated microscopy, and image analysis, an approach we call pharmacoscopy. First, the ability of pharmacoscopy to separate responders from non-responders was evaluated retrospectively for a cohort of 20 newly diagnosed and previously untreated patients with acute myeloid leukaemia. Next, 48 patients with aggressive haematological malignancies were prospectively evaluated for pharmacoscopy-guided treatment, of whom 17 could receive the treatment. The primary endpoint was progression-free survival in pharmacoscopy-treated patients, as compared with their own progression-free survival for the most recent regimen on which they had progressive disease. This trial is ongoing and registered with ClinicalTrials.gov, number NCT03096821. FINDINGS: Pharmacoscopy retrospectively predicted the clinical response of 20 acute myeloid leukaemia patients to initial therapy with 88·1% accuracy. In this interim analysis, 15 (88%) of 17 patients receiving pharmacoscopy-guided treatment had an overall response compared with four (24%) of 17 patients with their most recent regimen (odds ratio 24·38 [95% CI 3·99-125·4], p=0·0013). 12 (71%) of 17 patients had a progression-free survival ratio of 1·3 or higher, and median progression-free survival increased by four times, from 5·7 (95% CI 4·1-12·1) weeks to 22·6 (7·4-34·0) weeks (hazard ratio 3·14 [95% CI 1·37-7·22], p=0·0075). INTERPRETATION: Routine clinical integration of pharmacoscopy for treatment selection is technically feasible, and led to improved treatment of patients with aggressive refractory haematological malignancies in an initial patient cohort, warranting further investigation. FUNDING: Austrian Academy of Sciences; European Research Council; Austrian Science Fund; Austrian Federal Ministry of Science, Research and Economy; National Foundation for Research, Technology and Development; Anniversary Fund of the Austrian National Bank; MPN Research Foundation; European Molecular Biology Organization; and Swiss National Science Foundation.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Adenine/analogs & derivatives , Adult , Aged , Area Under Curve , Bone Marrow/pathology , Bortezomib/therapeutic use , Cladribine/therapeutic use , Disease-Free Survival , Female , Hematologic Neoplasms/diagnostic imaging , Hematologic Neoplasms/mortality , Hematologic Neoplasms/pathology , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Male , Microscopy, Fluorescence , Middle Aged , Odds Ratio , Pilot Projects , Piperidines , Positron Emission Tomography Computed Tomography , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , ROC Curve , Remission Induction , Young Adult
11.
Nat Chem Biol ; 13(6): 681-690, 2017 06.
Article in English | MEDLINE | ID: mdl-28437395

ABSTRACT

Small-molecule drugs may complement antibody-based therapies in an immune-oncology setting, yet systematic methods for the identification and characterization of the immunomodulatory properties of these entities are lacking. We surveyed the immumomodulatory potential of 1,402 small chemical molecules, as defined by their ability to alter the cell-cell interactions among peripheral mononuclear leukocytes ex vivo, using automated microscopy and population-wide single-cell image analysis. Unexpectedly, ∼10% of the agents tested affected these cell-cell interactions differentially. The results accurately recapitulated known immunomodulatory drug classes and revealed several clinically approved drugs that unexpectedly harbor the ability to modulate the immune system, which could potentially contribute to their physiological mechanism of action. For instance, the kinase inhibitor crizotinib promoted T cell interactions with monocytes, as well as with cancer cells, through inhibition of the receptor tyrosine kinase MSTR1 and subsequent upregulation of the expression of major histocompatibility complex molecules. The approach offers an attractive platform for the personalized identification and characterization of immunomodulatory therapeutics.


Subject(s)
Immunomodulation/drug effects , Small Molecule Libraries/pharmacology , Cardiac Myosins/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Crizotinib , Humans , Myosin Heavy Chains/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology
12.
Mol Cell Proteomics ; 15(3): 1139-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26933192

ABSTRACT

Tandem affinity purification-mass spectrometry (TAP-MS) is a popular strategy for the identification of protein-protein interactions, characterization of protein complexes, and entire networks. Its employment in cellular settings best fitting the relevant physiology is limited by convenient expression vector systems. We developed an easy-to-handle, inducible, dually selectable retroviral expression vector allowing dose- and time-dependent control of bait proteins bearing the efficient streptavidin-hemagglutinin (SH)-tag at their N- or C termini. Concomitant expression of a reporter fluorophore allows to monitor bait-expressing cells by flow cytometry or microscopy and enables high-throughput phenotypic assays. We used the system to successfully characterize the interactome of the neuroblastoma RAS viral oncogene homolog (NRAS) Gly12Asp (G12D) mutant and exploited the advantage of reporter fluorophore expression by tracking cytokine-independent cell growth using flow cytometry. Moreover, we tested the feasibility of studying cytotoxicity-mediating proteins with the vector system on the cell death-inducing mixed lineage kinase domain-like protein (MLKL) Ser358Asp (S358D) mutant. Interaction proteomics analysis of MLKL Ser358Asp (S358D) identified heat shock protein 90 (HSP90) as a high-confidence interacting protein. Further phenotypic characterization established MLKL as a novel HSP90 client. In summary, this novel inducible expression system enables SH-tag-based interaction studies in the cell line proficient for the respective phenotypic or signaling context and constitutes a valuable tool for experimental approaches requiring inducible or traceable protein expression.


Subject(s)
Chromatography, Affinity/methods , HSP90 Heat-Shock Proteins/metabolism , Mutation , Protein Kinases/metabolism , Proteomics/methods , Retroviridae/genetics , Tandem Mass Spectrometry/methods , Animals , Cell Line , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Genes, Reporter , HEK293 Cells , HT29 Cells , Humans , K562 Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Kinases/genetics
13.
J Biol Chem ; 291(3): 1123-36, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26555265

ABSTRACT

Many immunostimulants act as vaccine adjuvants via activation of the innate immune system, although in many cases it is unclear which specific molecules contribute to the stimulatory activity. QS-21 is a defined, highly purified, and soluble saponin adjuvant currently used in licensed and exploratory vaccines, including vaccines against malaria, cancer, and HIV-1. However, little is known about the mechanisms of cellular activation induced by QS-21. We observed QS-21 to elicit caspase-1-dependent IL-1ß and IL-18 release in antigen-presenting cells such as macrophages and dendritic cells when co-stimulated with the TLR4-agonist adjuvant monophosphoryl lipid A. Furthermore, our data suggest that the ASC-NLRP3 inflammasome is responsible for QS-21-induced IL-1ß/IL-18 release. At higher concentrations, QS-21 induced macrophage and dendritic cell death in a caspase-1-, ASC-, and NLRP3-independent manner, whereas the presence of cholesterol rescued cell viability. A nanoparticulate adjuvant that contains QS-21 as part of a heterogeneous mixture of saponins also induced IL-1ß in an NLRP3-dependent manner. Interestingly, despite the role NLRP3 plays for cellular activation in vitro, NLRP3-deficient mice immunized with HIV-1 gp120 and QS-21 showed significantly higher levels of Th1 and Th2 antigen-specific T cell responses and increased IgG1 and IgG2c compared with wild type controls. Thus, we have identified QS-21 as a nonparticulate single molecular saponin that activates the NLRP3 inflammasome, but this signaling pathway may contribute to decreased antigen-specific responses in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Carrier Proteins/metabolism , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Inflammasomes/drug effects , Macrophages/drug effects , Saponins/pharmacology , AIDS Vaccines/agonists , AIDS Vaccines/immunology , Adjuvants, Immunologic/analysis , Adjuvants, Immunologic/chemistry , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Carrier Proteins/genetics , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , HIV Envelope Protein gp120/agonists , HIV Envelope Protein gp120/immunology , Immunoglobulin G/analysis , Immunoglobulin G/biosynthesis , Inflammasomes/immunology , Inflammasomes/metabolism , Lipid A/agonists , Lipid A/analogs & derivatives , Lipid A/pharmacology , Macrophages/cytology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Saponins/analysis , Saponins/chemistry , Solubility , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
14.
NPJ Syst Biol Appl ; 2: 16027, 2016.
Article in English | MEDLINE | ID: mdl-28725479

ABSTRACT

Studying the relationship between virus infection and cellular response is paradigmatic for our understanding of how perturbation changes biological systems. Immune response, in this context is a complex yet evolutionarily adapted and robust cellular change, and is experimentally amenable to molecular analysis. To visualize the full cellular response to virus infection, we performed temporal transcriptomics, proteomics, and phosphoproteomics analysis of vesicular stomatitis virus (VSV)-infected mouse macrophages. This enabled the understanding of how infection-induced changes in host gene and protein expression are coordinated with post-translational modifications by cells in time to best measure and control the infection process. The vast and complex molecular changes measured could be decomposed in a limited number of clusters within each category (transcripts, proteins, and protein phosphorylation) each with own kinetic parameter and characteristic pathways/processes, suggesting multiple regulatory options in the overall sensing and homeostatic program. Altogether, the data underscored a prevalent executive function to phosphorylation. Resolution of the molecular events affecting the RIG-I pathway, central to viral recognition, reveals that phosphorylation of the key innate immunity adaptor mitochondrial antiviral-signaling protein (MAVS) on S328/S330 is necessary for activation of type-I interferon and nuclear factor κ B (NFκB) pathways. To further understand the hierarchical relationships, we analyzed kinase-substrate relationships and found RAF1 and, to a lesser extent, ARAF to be inhibiting VSV replication and necessary for NFκB activation, and AKT2, but not AKT1, to be supporting VSV replication. Integrated analysis using the omics data revealed co-regulation of transmembrane transporters including SLC7A11, which was subsequently validated as a host factor in the VSV replication. The data sets are predicted to greatly empower future studies on the functional organization of the response of macrophages to viral challenges.

15.
Cell ; 162(1): 170-83, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095250

ABSTRACT

Lipid composition affects the biophysical properties of membranes that provide a platform for receptor-mediated cellular signaling. To study the regulatory role of membrane lipid composition, we combined genetic perturbations of sphingolipid metabolism with the quantification of diverse steps in Toll-like receptor (TLR) signaling and mass spectrometry-based lipidomics. Membrane lipid composition was broadly affected by these perturbations, revealing a circular network of coregulated sphingolipids and glycerophospholipids. This evolutionarily conserved network architecture simultaneously reflected membrane lipid metabolism, subcellular localization, and adaptation mechanisms. Integration of the diverse TLR-induced inflammatory phenotypes with changes in lipid abundance assigned distinct functional roles to individual lipid species organized across the network. This functional annotation accurately predicted the inflammatory response of cells derived from patients suffering from lipid storage disorders, based solely on their altered membrane lipid composition. The analytical strategy described here empowers the understanding of higher-level organization of membrane lipid function in diverse biological systems.


Subject(s)
Immunity, Innate , Lipids/immunology , Animals , Cell Membrane/chemistry , Fibroblasts/metabolism , Gaucher Disease/immunology , Humans , Interleukin-6/immunology , Leukodystrophy, Globoid Cell/immunology , Metabolic Networks and Pathways , Mice , Sphingolipids/metabolism , Toll-Like Receptors/immunology
16.
Eur J Immunol ; 45(5): 1500-11, 2015 May.
Article in English | MEDLINE | ID: mdl-25678110

ABSTRACT

Secretion of type I interferon (IFN) is the first cellular reaction to invading pathogens. Despite the protective function of these cytokines, an excessive response to their action can contribute to serious pathologies, such as autoimmune diseases. Transcripts of most cytokines contain adenylate-uridylate (A/U)-rich elements (AREs) that make them highly unstable. RNA-binding proteins (RBPs) are mediators of the regulatory mechanisms that determine the fate of mRNAs containing AREs. Here, we applied an affinity proteomic approach and identified lethal, abnormal vision, drosophila-like 1 (ELAVL1)/Hu antigen R (HuR) as the predominant RBP of the IFN-ß mRNA ARE. Reduced expression or chemical inhibition of HuR severely hampered the type I IFN response in various cell lines and fibroblast-like synoviocytes isolated from joints of rheumatoid arthritis patients. These results define a role for HuR as a potent modulator of the type I IFN response. Taken together, HuR could be used as therapeutic target for diseases where type I IFN production is exaggerated.


Subject(s)
ELAV Proteins/immunology , Interferon Type I/biosynthesis , Interferon-beta/genetics , AU Rich Elements , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Base Sequence , ELAV Proteins/antagonists & inhibitors , ELAV Proteins/genetics , ELAV-Like Protein 1 , HeLa Cells , Humans , Interferon Inducers/pharmacology , Molecular Sequence Data , Poly I-C/pharmacology , Protein Multimerization , RNA Processing, Post-Transcriptional/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Synovial Membrane/immunology
17.
J Biol Chem ; 290(6): 3209-22, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25505250

ABSTRACT

Toll-like receptors (TLRs) are involved in sensing invading microbes by host innate immunity. TLR2 recognizes bacterial lipoproteins/lipopeptides, and lipopolysaccharide activates TLR4. TLR2 and TLR4 signal via the Toll/interleukin-1 receptor adaptors MyD88 and MAL, leading to NF-κB activation. TLR4 also utilizes the adaptors TRAM and TRIF, resulting in activation of interferon regulatory factor (IRF) 3. Here, we report a new role for TRAM and TRIF in TLR2 regulation and signaling. Interestingly, we observed that TLR2-mediated induction of the chemokine Ccl5 was impaired in TRAM or TRIF deficient macrophages. Inhibition of endocytosis reduced Ccl5 release, and the data also suggested that TRAM and TLR2 co-localize in early endosomes, supporting the hypothesis that signaling may occur from an intracellular compartment. Ccl5 release following lipoprotein challenge additionally involved the kinase Tbk-1 and Irf3, as well as MyD88 and Irf1. Induction of Interferon-ß and Ccl4 by lipoproteins was also partially impaired in cells lacking TRIF cells. Our results show a novel function of TRAM and TRIF in TLR2-mediated signal transduction, and the findings broaden our understanding of how Toll/interleukin-1 receptor adaptor proteins may participate in signaling downstream from TLR2.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cells, Cultured , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Endocytosis , Endosomes/metabolism , HEK293 Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin/genetics , Toll-Like Receptor 2/agonists
18.
Ann Rheum Dis ; 74(11): 2062-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24914072

ABSTRACT

OBJECTIVE: Nephrogenic systemic fibrosis (NSF) is a progressive fibrosing disorder that may develop in patients with chronic kidney disease after administration of gadolinium (Gd)-based contrast agents (GBCAs). In the setting of impaired renal clearance of GBCAs, Gd deposits in various tissues and fibrosis subsequently develops. However, the precise mechanism by which fibrosis occurs in NSF is incompletely understood. Because other profibrotic agents, such as silica or asbestos, activate the nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3) inflammasome and initiate interleukin (IL)-1ß release with the subsequent development of fibrosis, we evaluated the effects of GBCAs on inflammasome activation. METHODS: Bone marrow derived macrophages from C57BL/6, Nlrp3(-/-) and Asc(-/-) mice were incubated with three Gd-containing compounds and IL-1ß activation and secretion was detected by ELISA and western blot analysis. Inflammasome activation and regulation was investigated in IL-4- and interferon (IFN)γ-polarised macrophages by ELISA, quantitative real time (qRT)-PCR and NanoString nCounter analysis. Furthermore, C57BL/6 and Nlrp3(-/-)mice were intraperitoneally injected with GBCA and recruitment of inflammatory cells to the peritoneum was analysed by fluorescence-activated cell sorting (FACS). RESULTS: Free Gd and GBCAs activate the NLRP3 inflammasome and induce IL-1ß secretion in vitro. Gd-diethylenetriaminepentaacetic acid also induces the recruitment of neutrophils and inflammatory monocytes to the peritoneum in vivo. Gd activated IL-4-polarised macrophages more effectively than IFNγ-polarised macrophages, which preferentially expressed genes known to downregulate inflammasome activity. CONCLUSIONS: These data suggest that Gd released from GBCAs triggers a NLRP3 inflammasome-dependent inflammatory response that leads to fibrosis in an appropriate clinical setting. The preferential activation of IL-4-differentiated macrophages is consistent with the predominantly fibrotic presentation of NSF.


Subject(s)
Carrier Proteins/drug effects , Contrast Media/pharmacology , Gadolinium/pharmacology , Inflammasomes/drug effects , Interleukin-1beta/drug effects , Macrophages/drug effects , Nephrogenic Fibrosing Dermopathy/immunology , Peritoneum/drug effects , Peritonitis/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/immunology , Contrast Media/adverse effects , Disease Models, Animal , Gadolinium/adverse effects , Gadolinium DTPA/adverse effects , Gadolinium DTPA/pharmacology , Inflammasomes/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nephrogenic Fibrosing Dermopathy/chemically induced , Nephrogenic Fibrosing Dermopathy/genetics , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacology , Peritoneum/immunology , Peritonitis/chemically induced , Peritonitis/genetics
19.
Nat Commun ; 5: 5360, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406581

ABSTRACT

Primary immunodeficiency disorders enable identification of genes with crucial roles in the human immune system. Here we study patients suffering from recurrent bacterial, viral and Cryptosporidium infections, and identify a biallelic mutation in the MAP3K14 gene encoding NIK (NF-κB-inducing kinase). Loss of kinase activity of mutant NIK, predicted by in silico analysis and confirmed by functional assays, leads to defective activation of both canonical and non-canonical NF-κB signalling. Patients with mutated NIK exhibit B-cell lymphopenia, decreased frequencies of class-switched memory B cells and hypogammaglobulinemia due to impaired B-cell survival, and impaired ICOSL expression. Although overall T-cell numbers are normal, both follicular helper and memory T cells are perturbed. Natural killer (NK) cells are decreased and exhibit defective activation, leading to impaired formation of NK-cell immunological synapses. Collectively, our data illustrate the non-redundant role for NIK in human immune responses, demonstrating that loss-of-function mutations in NIK can cause multiple aberrations of lymphoid immunity.


Subject(s)
Agammaglobulinemia/genetics , Lymphopenia/genetics , Protein Serine-Threonine Kinases/genetics , Agammaglobulinemia/immunology , B-Lymphocytes/immunology , Bacterial Infections/immunology , Child, Preschool , Computer Simulation , Cryptosporidiosis/immunology , Female , Humans , Immunoglobulin Class Switching , Immunologic Deficiency Syndromes/genetics , Immunologic Memory , Inducible T-Cell Co-Stimulator Ligand/metabolism , Infant , Killer Cells, Natural/immunology , Leukocyte Count , Lymphocyte Count , Lymphopenia/immunology , Mutation , Pedigree , Recurrence , T-Lymphocytes, Helper-Inducer/immunology , Virus Diseases/immunology , NF-kappaB-Inducing Kinase
20.
Proc Natl Acad Sci U S A ; 111(20): 7391-6, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799678

ABSTRACT

A number of pathogens cause host cell death upon infection, and Yersinia pestis, infamous for its role in large pandemics such as the "Black Death" in medieval Europe, induces considerable cytotoxicity. The rapid killing of macrophages induced by Y. pestis, dependent upon type III secretion system effector Yersinia outer protein J (YopJ), is minimally affected by the absence of caspase-1, caspase-11, Fas ligand, and TNF. Caspase-8 is known to mediate apoptotic death in response to infection with several viruses and to regulate programmed necrosis (necroptosis), but its role in bacterially induced cell death is poorly understood. Here we provide genetic evidence for a receptor-interacting protein (RIP) kinase-caspase-8-dependent macrophage apoptotic death pathway after infection with Y. pestis, influenced by Toll-like receptor 4-TIR-domain-containing adapter-inducing interferon-ß (TLR4-TRIF). Interestingly, macrophages lacking either RIP1, or caspase-8 and RIP3, also had reduced infection-induced production of IL-1ß, IL-18, TNF, and IL-6; impaired activation of the transcription factor NF-κB; and greatly compromised caspase-1 processing. Cleavage of the proform of caspase-1 is associated with triggering inflammasome activity, which leads to the maturation of IL-1ß and IL-18, cytokines important to host responses against Y. pestis and many other infectious agents. Our results identify a RIP1-caspase-8/RIP3-dependent caspase-1 activation pathway after Y. pestis challenge. Mice defective in caspase-8 and RIP3 were also highly susceptible to infection and displayed reduced proinflammatory cytokines and myeloid cell death. We propose that caspase-8 and the RIP kinases are key regulators of macrophage cell death, NF-κB and inflammasome activation, and host resistance after Y. pestis infection.


Subject(s)
Caspase 8/metabolism , Cell Death , Immunity, Innate , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Bacterial Proteins/genetics , Bone Marrow Cells/cytology , Cytokines/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Yersinia Infections/microbiology , Yersinia pestis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...