Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 610(7930): 161-172, 2022 10.
Article in English | MEDLINE | ID: mdl-36171284

ABSTRACT

Expansion and differentiation of antigen-experienced PD-1+TCF-1+ stem-like CD8+ T cells into effector cells is critical for the success of immunotherapies based on PD-1 blockade1-4. Hashimoto et al. have shown that, in chronic infections, administration of the cytokine interleukin (IL)-2 triggers an alternative differentiation path of stem-like T cells towards a distinct population of 'better effector' CD8+ T cells similar to those generated in an acute infection5. IL-2 binding to the IL-2 receptor α-chain (CD25) was essential in triggering this alternative differentiation path and expanding better effectors with distinct transcriptional and epigenetic profiles. However, constitutive expression of CD25 on regulatory T cells and some endothelial cells also contributes to unwanted systemic effects from IL-2 therapy. Therefore, engineered IL-2 receptor ß- and γ-chain (IL-2Rßγ)-biased agonists are currently being developed6-10. Here we show that IL-2Rßγ-biased agonists are unable to preferentially expand better effector T cells in cancer models and describe PD1-IL2v, a new immunocytokine that overcomes the need for CD25 binding by docking in cis to PD-1. Cis binding of PD1-IL2v to PD-1 and IL-2Rßγ on the same cell recovers the ability to differentiate stem-like CD8+ T cells into better effectors in the absence of CD25 binding in both chronic infection and cancer models and provides superior efficacy. By contrast, PD-1- or PD-L1-blocking antibodies alone, or their combination with clinically relevant doses of non-PD-1-targeted IL2v, cannot expand this unique subset of better effector T cells and instead lead to the accumulation of terminally differentiated, exhausted T cells. These findings provide the basis for the development of a new generation of PD-1 cis-targeted IL-2R agonists with enhanced therapeutic potential for the treatment of cancer and chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Receptors, Interleukin-2 , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Infections/drug therapy , Infections/immunology , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/agonists , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Interleukin-2/agonists
2.
Nature ; 567(7746): 109-112, 2019 03.
Article in English | MEDLINE | ID: mdl-30787439

ABSTRACT

Zoonotic influenza A viruses of avian origin can cause severe disease in individuals, or even global pandemics, and thus pose a threat to human populations. Waterfowl and shorebirds are believed to be the reservoir for all influenza A viruses, but this has recently been challenged by the identification of novel influenza A viruses in bats1,2. The major bat influenza A virus envelope glycoprotein, haemagglutinin, does not bind the canonical influenza A virus receptor, sialic acid or any other glycan1,3,4, despite its high sequence and structural homology with conventional haemagglutinins. This functionally uncharacterized plasticity of the bat influenza A virus haemagglutinin means the tropism and zoonotic potential of these viruses has not been fully determined. Here we show, using transcriptomic profiling of susceptible versus non-susceptible cells in combination with genome-wide CRISPR-Cas9 screening, that the major histocompatibility complex class II (MHC-II) human leukocyte antigen DR isotype (HLA-DR) is an essential entry determinant for bat influenza A viruses. Genetic ablation of the HLA-DR α-chain rendered cells resistant to infection by bat influenza A virus, whereas ectopic expression of the HLA-DR complex in non-susceptible cells conferred susceptibility. Expression of MHC-II from different bat species, pigs, mice or chickens also conferred susceptibility to infection. Notably, the infection of mice with bat influenza A virus resulted in robust virus replication in the upper respiratory tract, whereas mice deficient for MHC-II were resistant. Collectively, our data identify MHC-II as a crucial entry mediator for bat influenza A viruses in multiple species, which permits a broad vertebrate tropism.


Subject(s)
Chiroptera/virology , Histocompatibility Antigens Class II/metabolism , Host Specificity , Influenza A virus/immunology , Influenza A virus/physiology , Zoonoses/immunology , Zoonoses/virology , Animals , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Chickens/genetics , Chickens/immunology , Chiroptera/genetics , Chiroptera/immunology , Chiroptera/metabolism , Female , Gene Expression Profiling , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Host Specificity/genetics , Host Specificity/immunology , Humans , Male , Mice , Mice, Knockout , Respiratory System/virology , Swine/genetics , Swine/immunology , Viral Tropism/genetics , Viral Tropism/immunology , Virus Replication , Zoonoses/genetics , Zoonoses/metabolism
4.
J Virol ; 89(1): 863-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320322

ABSTRACT

Human interferon-inducible transmembrane proteins (IFITMs) were identified as restriction factors of influenza A virus (IAV). Given the important role of pigs in the zoonotic cycle of IAV, we cloned swine IFITMs (swIFITMs) and found two IFITM1-like proteins, one homologue of IFITM2, and a homologue of IFITM3. We show that swIFITM2 and swIFITM3 localize to endosomes and display potent antiviral activities. Knockdown of swIFITMs strongly reduced virus inhibition by interferon, establishing the swIFITMs as potent restriction factors in porcine cells.


Subject(s)
Influenza A virus/immunology , Influenza A virus/physiology , Interferons/immunology , Membrane Proteins/immunology , Virus Replication , Animals , Cell Line , Endosomes/chemistry , Membrane Proteins/analysis , Swine
5.
PLoS Pathog ; 9(10): e1003632, 2013.
Article in English | MEDLINE | ID: mdl-24137104

ABSTRACT

Upon viral infection, the production of type I interferon (IFN) and the subsequent upregulation of IFN stimulated genes (ISGs) generate an antiviral state with an important role in the activation of innate and adaptive host immune responses. The ubiquitin-like protein (UBL) ISG15 is a critical IFN-induced antiviral molecule that protects against several viral infections, but the mechanism by which ISG15 exerts its antiviral function is not completely understood. Here, we report that ISG15 plays an important role in the regulation of macrophage responses. ISG15-/- macrophages display reduced activation, phagocytic capacity and programmed cell death activation in response to vaccinia virus (VACV) infection. Moreover, peritoneal macrophages from mice lacking ISG15 are neither able to phagocyte infected cells nor to block viral infection in co-culture experiments with VACV-infected murine embryonic fibroblast (MEFs). This phenotype is independent of cytokine production and secretion, but clearly correlates with impaired activation of the protein kinase AKT in ISG15 knock-out (KO) macrophages. Altogether, these results indicate an essential role of ISG15 in the cellular immune antiviral response and point out that a better understanding of the antiviral responses triggered by ISG15 may lead to the development of therapies against important human pathogens.


Subject(s)
Cytokines/metabolism , Immunity, Innate , Macrophages, Peritoneal/metabolism , Vaccinia virus/metabolism , Vaccinia/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/virology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitins/genetics , Ubiquitins/immunology , Ubiquitins/metabolism , Vaccinia/genetics , Vaccinia/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
6.
Med ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38593812

ABSTRACT

BACKGROUND: The treatment of melanoma, the deadliest form of skin cancer, has greatly benefited from immunotherapy. However, many patients do not show a durable response, which is only partially explained by known resistance mechanisms. METHODS: We performed single-cell RNA sequencing of tumor immune infiltrates and matched peripheral blood mononuclear cells of 22 checkpoint inhibitor (CPI)-naive stage III-IV metastatic melanoma patients. After sample collection, the same patients received CPI treatment, and their response was assessed. FINDINGS: CPI responders showed high levels of classical monocytes in peripheral blood, which preferentially transitioned toward CXCL9-expressing macrophages in tumors. Trajectories of tumor-infiltrating CD8+ T cells diverged at the level of effector memory/stem-like T cells, with non-responder cells progressing into a state characterized by cellular stress and apoptosis-related gene expression. Consistently, predicted non-responder-enriched myeloid-T/natural killer cell interactions were primarily immunosuppressive, while responder-enriched interactions were supportive of T cell priming and effector function. CONCLUSIONS: Our study illustrates that the tumor immune microenvironment prior to CPI treatment can be indicative of response. In perspective, modulating the myeloid and/or effector cell compartment by altering the described cell interactions and transitions could improve immunotherapy response. FUNDING: This research was funded by Roche Pharma Research and Early Development.

7.
NPJ Precis Oncol ; 8(1): 10, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38200223

ABSTRACT

The consensus molecular subtypes (CMS) of colorectal cancer (CRC) is the most widely-used gene expression-based classification and has contributed to a better understanding of disease heterogeneity and prognosis. Nevertheless, CMS intratumoral heterogeneity restricts its clinical application, stressing the necessity of further characterizing the composition and architecture of CRC. Here, we used Spatial Transcriptomics (ST) in combination with single-cell RNA sequencing (scRNA-seq) to decipher the spatially resolved cellular and molecular composition of CRC. In addition to mapping the intratumoral heterogeneity of CMS and their microenvironment, we identified cell communication events in the tumor-stroma interface of CMS2 carcinomas. This includes tumor growth-inhibiting as well as -activating signals, such as the potential regulation of the ETV4 transcriptional activity by DCN or the PLAU-PLAUR ligand-receptor interaction. Our study illustrates the potential of ST to resolve CRC molecular heterogeneity and thereby help advance personalized therapy.

8.
Int J Mol Sci ; 14(3): 4670-83, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23443165

ABSTRACT

Plants have developed versatile strategies to deal with the great variety of challenging conditions they are exposed to. Among them, the regulation of translation is a common target to finely modulate gene expression both under biotic and abiotic stress situations. Upon environmental challenges, translation is regulated to reduce the consumption of energy and to selectively synthesize proteins involved in the proper establishment of the tolerance response. In the case of viral infections, the situation is more complex, as viruses have evolved unconventional mechanisms to regulate translation in order to ensure the production of the viral encoded proteins using the plant machinery. Although the final purpose is different, in some cases, both plants and viruses share common mechanisms to modulate translation. In others, the mechanisms leading to the control of translation are viral- or stress-specific. In this paper, we review the different mechanisms involved in the regulation of translation initiation under virus infection and under environmental stress in plants. In addition, we describe the main features within the viral RNAs and the cellular mRNAs that promote their selective translation in plants undergoing biotic and abiotic stress situations.

9.
Clin Cancer Res ; 29(21): 4449-4463, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37379429

ABSTRACT

PURPOSE: Target-dependent TCB activity can result in the strong and systemic release of cytokines that may develop into cytokine release syndrome (CRS), highlighting the need to understand and prevent this complex clinical syndrome. EXPERIMENTAL DESIGN: We explored the cellular and molecular players involved in TCB-mediated cytokine release by single-cell RNA-sequencing of whole blood treated with CD20-TCB together with bulk RNA-sequencing of endothelial cells exposed to TCB-induced cytokine release. We used the in vitro whole blood assay and an in vivo DLBCL model in immunocompetent humanized mice to assess the effects of dexamethasone, anti-TNFα, anti-IL6R, anti-IL1R, and inflammasome inhibition, on TCB-mediated cytokine release and antitumor activity. RESULTS: Activated T cells release TNFα, IFNγ, IL2, IL8, and MIP-1ß, which rapidly activate monocytes, neutrophils, DCs, and NKs along with surrounding T cells to amplify the cascade further, leading to TNFα, IL8, IL6, IL1ß, MCP-1, MIP-1α, MIP-1ß, and IP-10 release. Endothelial cells contribute to IL6 and IL1ß release and at the same time release several chemokines (MCP-1, IP-10, MIP-1α, and MIP-1ß). Dexamethasone and TNFα blockade efficiently reduced CD20-TCB-mediated cytokine release whereas IL6R blockade, inflammasome inhibition, and IL1R blockade induced a less pronounced effect. Dexamethasone, IL6R blockade, IL1R blockade, and the inflammasome inhibitor did not interfere with CD20-TCB activity, in contrast to TNFα blockade, which partially inhibited antitumor activity. CONCLUSIONS: Our work sheds new light on the cellular and molecular players involved in cytokine release driven by TCBs and provides a rationale for the prevention of CRS in patients treated with TCBs. See related commentary by Luri-Rey et al., p. 4320.


Subject(s)
Antibodies, Bispecific , Tumor Necrosis Factor-alpha , Humans , Mice , Animals , Chemokine CCL3 , Chemokine CCL4 , Antibodies, Bispecific/pharmacology , Interleukin-8 , Chemokine CXCL10 , Interleukin-6 , Cytokine Release Syndrome , Endothelial Cells , Inflammasomes , Cytokines , T-Lymphocytes , Dexamethasone/pharmacology , RNA
11.
Virol J ; 8: 147, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21450110

ABSTRACT

Duck hepatitis virus (DHV-1) is a worldwide distributed picornavirus that causes acute and fatal disease in young ducklings. Recently, the complete genome of DHV-1 has been determined and comparative sequence analysis has shown that possesses the typical picornavirus organization but exhibits several unique features. For the first time, we provide evidence that the 626-nucleotide-long 5'-UTR of the DHV-1 genome contains an internal ribosome entry site (IRES) element that functions efficiently both in vitro and in mammalian cells. The prediction of the secondary structure of the DHV-1 IRES shows significant similarity to the hepatitis C virus (HCV) IRES. Moreover, similarly to HCV IRES, DHV-1 IRES can direct translation initiation in the absence of a functional eIF4F complex. We also demonstrate that the activity of the DHV-1 IRES is modulated by a viral coding sequence located downstream of the DHV-1 5'-UTR, which enhances DHV-1 IRES activity both in vitro and in vivo. Furthermore, mutational analysis of the predicted pseudo-knot structures at the 3'-end of the putative DHV-1 IRES supported the presence of conserved domains II and III and, as it has been previously described for other picornaviruses, these structures are essential for keeping the normal internal initiation of translation of DHV-1.


Subject(s)
5' Untranslated Regions , Eukaryotic Initiation Factor-4F/metabolism , Gene Expression Regulation, Viral , Hepacivirus/genetics , Hepatitis Virus, Duck/genetics , Hepatitis, Viral, Animal/metabolism , Picornaviridae Infections/veterinary , Animals , Base Sequence , Cell Line , Chickens , Conserved Sequence , Cricetinae , Eukaryotic Initiation Factor-4F/genetics , Genome, Viral , Hepacivirus/chemistry , Hepatitis Virus, Duck/chemistry , Hepatitis Virus, Duck/metabolism , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/virology , Molecular Sequence Data , Nucleic Acid Conformation , Picornaviridae Infections/genetics , Picornaviridae Infections/metabolism , Picornaviridae Infections/virology , Protein Binding , Protein Biosynthesis , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism
12.
Elife ; 102021 04 23.
Article in English | MEDLINE | ID: mdl-33890853

ABSTRACT

Peripheral nerves are organ-like structures containing diverse cell types to optimize function. This interactive assembly includes mostly axon-associated Schwann cells, but also endothelial cells of supporting blood vessels, immune system-associated cells, barrier-forming cells of the perineurium surrounding and protecting nerve fascicles, and connective tissue-resident cells within the intra-fascicular endoneurium and inter-fascicular epineurium. We have established transcriptional profiles of mouse sciatic nerve-inhabitant cells to foster the fundamental understanding of peripheral nerves. To achieve this goal, we have combined bulk RNA sequencing of developing sciatic nerves up to the adult with focused bulk and single-cell RNA sequencing of Schwann cells throughout postnatal development, extended by single-cell transcriptome analysis of the full sciatic nerve both perinatally and in the adult. The results were merged in the transcriptome resource Sciatic Nerve ATlas (SNAT: https://www.snat.ethz.ch). We anticipate that insights gained from our multi-layered analysis will serve as valuable interactive reference point to guide future studies.


Subject(s)
Peripheral Nerves/metabolism , Transcription, Genetic , Animals , Female , Gene Expression Profiling , Male , Mice , Mice, Transgenic , Sciatic Nerve/metabolism
13.
Medicina (Kaunas) ; 46(6): 393-400, 2010.
Article in English | MEDLINE | ID: mdl-20944447

ABSTRACT

BACKGROUND: Hospitalization processes are known to increase depressive symptoms arising among elderly population. Meanwhile, dysregulation of cardiac autonomic function has been suggested to link depression and cardiovascular mortality. In this context, analysis of heart rate variability (HRV) is emerging as a powerful mortality risk stratifier clinical tool. The purpose of the study was to examine the relationship among HRV, depression, and comorbidity risk among an elderly inpatient population. MATERIAL AND METHODS: Twenty-six subjects (aged 78±9 years) were recruited from the Short-Term Stay Unit at the Hospital General de Alicante. Before joining a Physical Activity Program aimed to prevent functional impairment and after medical selection and written consent, inpatients were tested for heart rate variability, Yesavage Geriatric Depression Scale, and Charlson comorbidity index score. RESULTS: Men compared to women showed a significantly larger CCI score. Short-term scaling exponent (α(1)), derived from detrended fluctuation analysis, showed a negative correlation with Charlson comorbidity index. Conversely, a positive correlation was found between sample entropy (SampEn) and Yesavage Scale. CONCLUSIONS: On the one hand, fractal analysis of HRV confirms to be useful as a risk stratifier tool. On the other hand, SampEn is proposed to be reflecting a non-neurally generated complexity when accompanied with low values of α(1). Accordingly, in this regime, it would be indicative of a paradoxical gradual reduction in cardiac autonomic control, accentuated with the severity of depressive symptoms.


Subject(s)
Depression/epidemiology , Heart Diseases/epidemiology , Heart Rate/physiology , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Comorbidity , Data Interpretation, Statistical , Depression/complications , Depression/diagnosis , Depression/mortality , Electrocardiography , Female , Geriatric Assessment , Heart Diseases/mortality , Humans , Inpatients , Male , Nonlinear Dynamics , Prevalence , Risk Assessment , Sampling Studies , Sex Factors
14.
Virology ; 539: 104-113, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31706162

ABSTRACT

The vRNAs of influenza A viruses contain 12 and 13 nucleotide-long sequences at their 3' and 5' termini respectively that are highly conserved and constitute the vRNA promoter. These sequences and the next three segment-specific nucleotides show inverted partial complementarity and are followed by several unpaired nucleotides of poorly characterized function at the 3' end. We have performed systematic point-mutations at the segment-specific nucleotides 15-18 of the 3'-end of a NS-like vRNA segment. All NS-like vRNAs containing mutations at position 15, and some at positions 16-18 showed reduced transcription/replication efficiency in a transfection/infection system. In addition, the replication of recombinant viruses containing mutations at position 15 was impaired both in single and multi-cycle experiments. This reduction was the consequence of a decreased expression of the NS segment. The data indicate that NS1 plays a role in the transcription/replication of its own segment, which elicits a global defect on virus replication.


Subject(s)
3' Untranslated Regions/genetics , Influenza A virus/physiology , Virus Replication , A549 Cells , Animals , Dogs , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Mutation , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
15.
STAR Protoc ; 1(2): 100081, 2020 09 18.
Article in English | MEDLINE | ID: mdl-33000004

ABSTRACT

This protocol presents a plate-based workflow to perform RNA sequencing analysis of single cells/nuclei using Smart-seq2. We describe (1) the dissociation procedures for cell/nucleus isolation from the mouse brain and human organoids, (2) the flow sorting of single cells/nuclei into 384-well plates, and (3) the preparation of libraries following miniaturization of the Smart-seq2 protocol using a liquid-handling robot. This pipeline allows for the reliable, high-throughput, and cost-effective preparation of mouse and human samples for full-length deep single-cell/nucleus RNA sequencing. For complete details on the use and execution of this protocol, please refer to Bowers et al. (2020).


Subject(s)
Sequence Analysis, RNA/instrumentation , Single-Cell Analysis/instrumentation , Single-Cell Analysis/methods , Animals , Base Sequence/genetics , Brain/cytology , Brain/metabolism , Cell Nucleus/metabolism , Cell Separation/methods , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Library , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Miniaturization , RNA/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Exome Sequencing/methods , Workflow
16.
Methods Mol Biol ; 1836: 59-88, 2018.
Article in English | MEDLINE | ID: mdl-30151569

ABSTRACT

Influenza viruses are constantly circulating among humans, in which they cause seasonal epidemics of severe respiratory disease. Additionally, these zoonotic viruses infect different mammals and birds, from which new antigenic variants are occasionally transmitted to humans leading to devastating global pandemics. Surveillance programs, in which viruses from the main reservoir (waterfowl), intermediate hosts (like pigs and other farm animals), and other affected species are isolated and characterized, are crucial for the global influenza prevention strategy. This chapter gives an overview of the most commonly used methods for the propagation and titration of influenza viruses, which are key steps in surveillance procedures, as well as in vaccine development and basic research. Depending on the host and the viral strain, primary isolates are obtained from biological samples of different origin and subsequently amplified in embryonated chicken eggs or cell cultures. These propagation procedures are the focus of the first part of this chapter. Once the initial isolates have been amplified, virus titration methods based on particular characteristics of influenza viruses, such as their ability to agglutinate red blood cells (RBCs) or to induce cytopathic effects (CPE) in cell monolayers, are used to estimate the amount of viral particles. Such approaches, like the hemagglutination assay (HA assay), 50% tissue culture infectious dose (TCID50), or plaque assay, are included in the second part of this chapter. Although they are simple and cost-effective, some of these techniques have been partially replaced by faster and more sensitive methods based on the quantification of viral genomes, such as the quantitative real-time reverse transcription PCR (RT-qPCR), which is presented at the end of this section. The different protocols are explained in detail in order to facilitate the preparation and quantification of infectious virus stocks.


Subject(s)
Influenza, Human/diagnosis , Influenza, Human/virology , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/virology , Orthomyxoviridae/physiology , Viral Load , Virus Replication , Animals , Cell Line , Cells, Cultured , Chick Embryo , Hemagglutination Tests , Humans , Orthomyxoviridae/isolation & purification , Real-Time Polymerase Chain Reaction , Swine , Viral Plaque Assay
17.
Nat Commun ; 9(1): 3679, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30206219

ABSTRACT

Although annual influenza epidemics affect around 10% of the global population, current treatment options are limited and development of new antivirals is needed. Here, using quantitative phosphoproteomics, we reveal the unique phosphoproteome dynamics that occur in the host cell within minutes of influenza A virus (IAV) infection. We uncover cellular kinases required for the observed signaling pattern and find that inhibition of selected candidates, such as the G protein-coupled receptor kinase 2 (GRK2), leads to decreased IAV replication. As GRK2 has emerged as drug target in heart disease, we focus on its role in IAV infection and show that it is required for viral uncoating. Replication of seasonal and pandemic IAVs is severely decreased by specific GRK2 inhibitors in primary human airway cultures and in mice. Our study reveals the IAV-induced changes to the cellular phosphoproteome and identifies GRK2 as crucial node of the kinase network that enables IAV replication.


Subject(s)
Antiviral Agents/pharmacology , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Influenza, Human/metabolism , Influenza, Human/virology , Molecular Targeted Therapy , Phosphoproteins/metabolism , Protein Kinases/metabolism , Proteomics/methods , Amino Acid Sequence , Animals , Cells, Cultured , Enzyme Activation/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , Humans , Lung/pathology , Lung/virology , Mice , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Phosphoproteins/chemistry , Phosphorylation/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects
18.
Sci Rep ; 7(1): 8629, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819179

ABSTRACT

In recent years genome-wide RNAi screens have revealed hundreds of cellular factors required for influenza virus infections in human cells. The long-term goal is to establish some of them as drug targets for the development of the next generation of antivirals against influenza. We found that several members of the polo-like kinases (PLK), a family of serine/threonine kinases with well-known roles in cell cycle regulation, were identified as hits in four different RNAi screens and we therefore studied their potential as drug target for influenza. We show that knockdown of PLK1, PLK3, and PLK4, as well as inhibition of PLK kinase activity by four different compounds, leads to reduced influenza virus replication, and we map the requirement of PLK activity to early stages of the viral replication cycle. We also tested the impact of the PLK inhibitor BI2536 on influenza virus replication in a human lung tissue culture model and observed strong inhibition of virus replication with no measurable toxicity. This study establishes the PLKs as potential drug targets for influenza and contributes to a more detailed understanding of the intricate interactions between influenza viruses and their host cells.


Subject(s)
Influenza A virus/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Virus Replication/drug effects , A549 Cells , Animals , Antimitotic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dogs , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , Influenza A virus/physiology , Influenza, Human/prevention & control , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pteridines/pharmacology , RNA Interference , Sulfones/pharmacology , Tumor Suppressor Proteins , Polo-Like Kinase 1
19.
mBio ; 6(3): e00297, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26060270

ABSTRACT

UNLABELLED: Human cathepsin W (CtsW) is a cysteine protease, which was identified in a genome-wide RNA interference (RNAi) screen to be required for influenza A virus (IAV) replication. In this study, we show that reducing the levels of expression of CtsW reduces viral titers for different subtypes of IAV, and we map the target step of CtsW requirement to viral entry. Using a set of small interfering RNAs (siRNAs) targeting CtsW, we demonstrate that knockdown of CtsW results in a decrease of IAV nucleoprotein accumulation in the nuclei of infected cells at 3 h postinfection. Assays specific for the individual stages of IAV entry further show that attachment, internalization, and early endosomal trafficking are not affected by CtsW knockdown. However, we detected impaired escape of viral particles from late endosomes in CtsW knockdown cells. Moreover, fusion analysis with a dual-labeled influenza virus revealed a significant reduction in fusion events, with no detectable impact on endosomal pH, suggesting that CtsW is required at the stage of viral fusion. The defect in IAV entry upon CtsW knockdown could be rescued by ectopic expression of wild-type CtsW but not by the expression of a catalytically inactive mutant of CtsW, suggesting that the proteolytic activity of CtsW is required for successful entry of IAV. Our results establish CtsW as an important host factor for entry of IAV into target cells and suggest that CtsW could be a promising target for the development of future antiviral drugs. IMPORTANCE: Increasing levels of resistance of influenza viruses to available antiviral drugs have been observed. Development of novel treatment options is therefore of high priority. In parallel to the classical approach of targeting viral enzymes, a novel strategy is pursued: cell-dependent factors of the virus are identified with the aim of developing small-molecule inhibitors against a cellular target that the virus relies on. For influenza A virus, several genome-wide RNA interference (RNAi) screens revealed hundreds of potential cellular targets. However, we have only limited knowledge on how these factors support virus replication, which would be required for drug development. We have characterized cathepsin W, one of the candidate factors, and found that cathepsin W is required for escape of influenza virus from the late endosome. Importantly, this required the proteolytic activity of cathepsin W. We therefore suggest that cathepsin W could be a target for future host cell-directed antiviral therapies.


Subject(s)
Cathepsin W/metabolism , Endosomes/virology , Host-Pathogen Interactions , Influenza A virus/physiology , Virus Internalization , Animals , Cell Line , Gene Knockdown Techniques , Genetic Complementation Test , Genetic Testing , Humans
20.
Cell Host Microbe ; 18(6): 723-35, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26651948

ABSTRACT

Several systems-level datasets designed to dissect host-pathogen interactions during influenza A infection have been reported. However, apparent discordance among these data has hampered their full utility toward advancing mechanistic and therapeutic knowledge. To collectively reconcile these datasets, we performed a meta-analysis of data from eight published RNAi screens and integrated these data with three protein interaction datasets, including one generated within the context of this study. Further integration of these data with global virus-host interaction analyses revealed a functionally validated biochemical landscape of the influenza-host interface, which can be queried through a simplified and customizable web portal (http://www.metascape.org/IAV). Follow-up studies revealed that the putative ubiquitin ligase UBR4 associates with the viral M2 protein and promotes apical transport of viral proteins. Taken together, the integrative analysis of influenza OMICs datasets illuminates a viral-host network of high-confidence human proteins that are essential for influenza A virus replication.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Cytoskeletal Proteins/metabolism , Host-Pathogen Interactions , Influenza A virus/physiology , Viral Matrix Proteins/metabolism , Virus Release , Animals , Cell Line , Computational Biology , Flow Cytometry , Humans , Immunoprecipitation , Mice, Inbred BALB C , Microscopy, Fluorescence , Protein Binding , Protein Interaction Maps , Protein Transport , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL