Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nat Rev Mol Cell Biol ; 21(12): 750-764, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33093672

RESUMO

Cell-cell interfaces are found throughout multicellular organisms, from transient interactions between motile immune cells to long-lived cell-cell contacts in epithelia. Studies of immune cell interactions, epithelial cell barriers, neuronal contacts and sites of cell-cell fusion have identified a core set of features shared by cell-cell interfaces that critically control their function. Data from diverse cell types also show that cells actively and passively regulate the localization, strength, duration and cytoskeletal coupling of receptor interactions governing cell-cell signalling and physical connections between cells, indicating that cell-cell interfaces have a unique membrane organization that emerges from local molecular and cellular mechanics. In this Review, we discuss recent findings that support the emerging view of cell-cell interfaces as specialized compartments that biophysically constrain the arrangement and activity of their protein, lipid and glycan components. We also review how these biophysical features of cell-cell interfaces allow cells to respond with high selectivity and sensitivity to multiple inputs, serving as the basis for wide-ranging cellular functions. Finally, we consider how the unique properties of cell-cell interfaces present opportunities for therapeutic intervention.


Assuntos
Comunicação Celular/fisiologia , Compartimento Celular/fisiologia , Fenômenos Fisiológicos Celulares/fisiologia , Animais , Fusão Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Humanos , Mecanotransdução Celular/fisiologia , Neurônios/citologia , Neurônios/fisiologia
2.
Immunity ; 51(2): 310-323.e7, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31204070

RESUMO

The tumor necrosis factor receptor superfamily member HVEM is one of the most frequently mutated surface proteins in germinal center (GC)-derived B cell lymphomas. We found that HVEM deficiency increased B cell competitiveness during pre-GC and GC responses. The immunoglobulin (Ig) superfamily protein BTLA regulated HVEM-expressing B cell responses independently of B-cell-intrinsic signaling via HVEM or BTLA. BTLA signaling into T cells through the phosphatase SHP1 reduced T cell receptor (TCR) signaling and preformed CD40 ligand mobilization to the immunological synapse, thus diminishing the help delivered to B cells. Moreover, T cell deficiency in BTLA cooperated with B cell Bcl-2 overexpression, leading to GC B cell outgrowth. These results establish that HVEM restrains the T helper signals delivered to B cells to influence GC selection outcomes, and they suggest that BTLA functions as a cell-extrinsic suppressor of GC B cell lymphomagenesis.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Proliferação de Células , Sinapses Imunológicas , Ativação Linfocitária , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Comunicação Parácrina , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Imunológicos/genética , Transdução de Sinais
3.
Nature ; 587(7833): 309-312, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32650338

RESUMO

The Plasmodium species that cause malaria are obligate intracellular parasites, and disease symptoms occur when these parasites replicate in human blood. Despite the risk of immune detection, the parasite delivers proteins that bind to host receptors on the cell surfaces of infected erythrocytes. In the causative parasite of the most deadly form of malaria in humans, Plasmodium falciparum, RIFINs form the largest family of surface proteins displayed by erythrocytes1. Some RIFINs can bind to inhibitory immune receptors, and these RIFINs act as targets for unusual antibodies that contain a LAIR1 ectodomain2-4 or as ligands for LILRB15. RIFINs stimulate the activation of and signalling by LILRB15, which could potentially lead to the dampening of human immune responses. Here, to understand how RIFINs activate LILRB1-mediated signalling, we determine the structure of a RIFIN bound to LILRB1. We show that this RIFIN mimics the natural activating ligand of LILRB1, MHC class I, in its LILRB1-binding mode. A single mutation in the RIFIN disrupts the complex, blocks LILRB1 binding of all tested RIFINs and abolishes signalling in a reporter assay. In a supported lipid bilayer system, which mimics the activation of natural killer (NK) cells by antibody-dependent cell-mediated cytotoxicity, both RIFIN and MHC are recruited to the immunological synapse of NK cells and reduce the activation of NK cells, as measured by the mobilization of perforin. Therefore, LILRB1-binding RIFINs mimic the binding mode of the natural ligand of LILRB1 and suppress the function of NK cells.


Assuntos
Receptor B1 de Leucócitos Semelhante a Imunoglobulina/química , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/imunologia , Malária Falciparum/imunologia , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sítios de Ligação/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Receptor B1 de Leucócitos Semelhante a Imunoglobulina/metabolismo , Ligantes , Bicamadas Lipídicas , Ativação Linfocitária , Malária Falciparum/parasitologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mimetismo Molecular/imunologia , Mutação , Perforina/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Transdução de Sinais
4.
Biophys J ; 2023 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-37596785

RESUMO

Formation of the immunological synapse (IS) is a key event during initiation of an adaptive immune response to a specific antigen. During this process, a T cell and an antigen presenting cell form a stable contact that allows the T cell to integrate both internal and external stimuli in order to decide whether to activate. The threshold for T cell activation depends on the strength and frequency of the calcium (Ca2+) signaling induced by antigen recognition, and it must be tightly regulated to avoid undesired harm to healthy cells. Potassium (K+) channels are recruited to the IS to maintain the negative membrane potential required to sustain Ca2+ entry. However, the precise localization of K+ channels within the IS remains unknown. Here, we visualized the dynamic subsynaptic distribution of Kv1.3, the main voltage-gated potassium channel in human T cells. Upon T cell receptor engagement, Kv1.3 polarized toward the synaptic cleft and diffused throughout the F-actin rich distal compartment of the synaptic interface-an effect enhanced by CD2-CD58 corolla formation. As the synapse matured, Kv1.3 clusters were internalized at the center of the IS and released in extracellular vesicles. We propose a model in which specific distribution of Kv1.3 within the synapse indirectly regulates the channel function and that this process is limited through Kv1.3 internalization and release in extracellular vesicles.

5.
J Cell Sci ; 132(4)2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30209137

RESUMO

The spatiotemporal regulation of signalling proteins at the contacts formed between immune cells and their targets determines how and when immune responses begin and end. Therapeutic control of immune responses therefore relies on thorough elucidation of the molecular processes occurring at these interfaces. However, the detailed investigation of each component's contribution to the formation and regulation of the contact is hampered by the complexities of cell composition and architecture. Moreover, the transient nature of these interactions creates additional challenges, especially in the use of advanced imaging technology. One approach that circumvents these problems is to establish in vitro systems that faithfully mimic immune cell interactions, but allow complexity to be 'dialled-in' as needed. Here, we present an in vitro system that makes use of synthetic vesicles that mimic important aspects of immune cell surfaces. Using this system, we began to explore the spatial distribution of signalling molecules (receptors, kinases and phosphatases) and how this changes during the initiation of signalling. The GUV/cell system presented here is expected to be widely applicable.


Assuntos
Comunicação Celular/imunologia , Membrana Celular/metabolismo , Transdução de Sinais/imunologia , Lipossomas Unilamelares/metabolismo , Humanos , Células Jurkat , Fosfatidilcolinas/imunologia , Fosfatidilcolinas/metabolismo , Ligação Proteica/imunologia , Lipossomas Unilamelares/imunologia
6.
Biophys J ; 116(1): 31-41, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30558888

RESUMO

G-protein-coupled receptors (GPCRs) comprise the largest and most pharmacologically important family of cell-surface receptors encoded by the human genome. In many instances, the distinct signaling behavior of certain GPCRs has been explained in terms of the formation of heteromers with, for example, distinct signaling properties and allosteric cross-regulation. Confirmation of this has, however, been limited by the paucity of reliable methods for probing heteromeric GPCR interactions in situ. The most widely used assays for GPCR stoichiometry, based on resonance energy transfer, are unsuited to reporting heteromeric interactions. Here, we describe a targeted bioluminescence resonance energy transfer (BRET) assay, called type-4 BRET, which detects both homo- and heteromeric interactions using induced multimerization of protomers within such complexes, at constant expression. Using type-4 BRET assays, we investigate heterodimerization among known GPCR homodimers: the CXC chemokine receptor 4 and sphingosine-1-phosphate receptors. We observe that CXC chemokine receptor 4 and sphingosine-1-phosphate receptors can form heterodimers with GPCRs from their immediate subfamilies but not with more distantly related receptors. We also show that heterodimerization appears to disrupt homodimeric interactions, suggesting the sharing of interfaces. Broadly, these observations indicate that heterodimerization results from the divergence of homodimeric receptors and will therefore likely be restricted to closely related homodimeric GPCRs.


Assuntos
Multimerização Proteica , Receptores CXCR4/química , Receptores de Esfingosina-1-Fosfato/química , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Receptores de Esfingosina-1-Fosfato/metabolismo
7.
Nature ; 488(7412): 508-511, 2012 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-22801493

RESUMO

Although there has been much success in identifying genetic variants associated with common diseases using genome-wide association studies (GWAS), it has been difficult to demonstrate which variants are causal and what role they have in disease. Moreover, the modest contribution that these variants make to disease risk has raised questions regarding their medical relevance. Here we have investigated a single nucleotide polymorphism (SNP) in the TNFRSF1A gene, that encodes tumour necrosis factor receptor 1 (TNFR1), which was discovered through GWAS to be associated with multiple sclerosis (MS), but not with other autoimmune conditions such as rheumatoid arthritis, psoriasis and Crohn's disease. By analysing MS GWAS data in conjunction with the 1000 Genomes Project data we provide genetic evidence that strongly implicates this SNP, rs1800693, as the causal variant in the TNFRSF1A region. We further substantiate this through functional studies showing that the MS risk allele directs expression of a novel, soluble form of TNFR1 that can block TNF. Importantly, TNF-blocking drugs can promote onset or exacerbation of MS, but they have proven highly efficacious in the treatment of autoimmune diseases for which there is no association with rs1800693. This indicates that the clinical experience with these drugs parallels the disease association of rs1800693, and that the MS-associated TNFR1 variant mimics the effect of TNF-blocking drugs. Hence, our study demonstrates that clinical practice can be informed by comparing GWAS across common autoimmune diseases and by investigating the functional consequences of the disease-associated genetic variation.


Assuntos
Predisposição Genética para Doença/genética , Esclerose Múltipla/induzido quimicamente , Esclerose Múltipla/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Alelos , Éxons/genética , Genoma Humano/genética , Estudo de Associação Genômica Ampla , Genômica , Genótipo , Humanos , Esclerose Múltipla/tratamento farmacológico , Splicing de RNA/genética , Receptores Tipo I de Fatores de Necrose Tumoral/análise , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Solubilidade , Fator de Necrose Tumoral alfa/metabolismo , Reino Unido
8.
Nano Lett ; 16(4): 2633-8, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-26923775

RESUMO

Measuring small forces is a major challenge in cell biology. Here we improve the spatial resolution and accuracy of force reconstruction of the well-established technique of traction force microscopy (TFM) using STED microscopy. The increased spatial resolution of STED-TFM (STFM) allows a greater than 5-fold higher sampling of the forces generated by the cell than conventional TFM, accessing the nano instead of the micron scale. This improvement is highlighted by computer simulations and an activating RBL cell model system.


Assuntos
Simulação por Computador , Microscopia de Varredura por Sonda , Modelos Teóricos , Tração , Algoritmos , Adesão Celular , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Microscopia de Varredura por Sonda/instrumentação , Microscopia de Varredura por Sonda/métodos , Estresse Mecânico
9.
Biophys J ; 109(9): 1798-806, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26536257

RESUMO

The extent to which Rhodopsin family G-protein-coupled receptors (GPCRs) form invariant oligomers is contentious. Recent single-molecule fluorescence imaging studies mostly argue against the existence of constitutive receptor dimers and instead suggest that GPCRs only dimerize transiently, if at all. However, whether or not even transient dimers exist is not always clear due to difficulties in unambiguously distinguishing genuine interactions from chance colocalizations, particularly with respect to short-lived events. Previous single-molecule studies have depended critically on calculations of chance colocalization rates and/or comparison with unfixed control proteins whose diffusional behavior may or may not differ from that of the test receptor. Here, we describe a single-molecule imaging assay that 1) utilizes comparisons with well-characterized control proteins, i.e., the monomer CD86 and the homodimer CD28, and 2) relies on cell fixation to limit artifacts arising from differences in the distribution and diffusion of test proteins versus these controls. The improved assay reliably reports the stoichiometry of the Glutamate-family GPCR dimer, γ-amino butyric acid receptor b2, whereas two Rhodopsin-family GPCRs, ß2-adrenergic receptor and mCannR2, exhibit colocalization levels comparable to those of CD86 monomers, strengthening the case against invariant GPCR oligomerization.


Assuntos
Microscopia de Fluorescência/métodos , Receptores Adrenérgicos beta 2/metabolismo , Receptores de GABA-B/metabolismo , Animais , Artefatos , Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Células CHO , Cricetulus , Difusão , Dimerização , Células HEK293 , Humanos , Transfecção
10.
Biophys J ; 106(12): L41-3, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24940791

RESUMO

We show that in conventional, competition-based bioluminescence resonance energy transfer (BRET) assays of membrane protein stoichiometry, the presence of competitors can alter tagged-protein density and artifactually reduce energy transfer efficiency. A well-characterized monomeric type I membrane protein, CD86, and two G protein-coupled receptors ß2AR and mCannR2, all of which behave as dimers in these conventional assays, exhibit monomeric behavior in an improved competition-based type-3 BRET assay designed to circumvent such artifacts.


Assuntos
Transferência de Energia , Medições Luminescentes/métodos , Animais , Proteínas de Membrana/metabolismo , Camundongos , Receptores Acoplados a Proteínas G/metabolismo
11.
J Biol Chem ; 286(37): 31993-2001, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21757710

RESUMO

Understanding the component stoichiometry of the T cell antigen receptor (TCR) triggering apparatus is essential for building realistic models of signal initiation. Recent studies suggesting that the TCR and other signaling-associated proteins are preclustered on resting T cells relied on measurements of the behavior of membrane proteins at interfaces with functionalized glass surfaces. Using fluorescence recovery after photobleaching, we show that, compared with the apical surface, the mobility of TCRs is significantly reduced at Jurkat T cell/glass interfaces, in a signaling-sensitive manner. Using two biophysical approaches that mitigate these effects, bioluminescence resonance energy transfer and two-color coincidence detection microscopy, we show that, within the uncertainty of the methods, the membrane components of the TCR triggering apparatus, i.e. the TCR complex, MHC molecules, CD4/Lck and CD45, are exclusively monovalent or monomeric in human T cell lines, implying that TCR triggering depends only on the kinetics of TCR/pMHC interactions. These analyses also showed that constraining proteins to two dimensions at the cell surface greatly enhances random interactions versus those between the membrane and the cytoplasm. Simulations of TCR-pMHC complex formation based on these findings suggest how unclustered TCR triggering-associated proteins might nevertheless be capable of generating complex signaling outputs via the differential recruitment of cytosolic effectors to the cell membrane.


Assuntos
Antígenos CD4/imunologia , Antígenos HLA/imunologia , Antígenos Comuns de Leucócito/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Receptores de Antígenos/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Antígenos CD4/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Citosol/imunologia , Citosol/metabolismo , Células HEK293 , Antígenos HLA/metabolismo , Humanos , Células Jurkat , Antígenos Comuns de Leucócito/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Modelos Imunológicos , Receptores de Antígenos/metabolismo , Linfócitos T/metabolismo
12.
J Biol Chem ; 286(27): 24208-18, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21566123

RESUMO

The human C-type lectin-like molecule CLEC5A is a critical macrophage receptor for dengue virus. The binding of dengue virus to CLEC5A triggers signaling through the associated adapter molecule DAP12, stimulating proinflammatory cytokine release. We have crystallized an informative ensemble of CLEC5A structural conformers at 1.9-Å resolution and demonstrate how an on-off extension to a ß-sheet acts as a binary switch regulating the flexibility of the molecule. This structural information together with molecular dynamics simulations suggests a mechanism whereby extracellular events may be transmitted through the membrane and influence DAP12 signaling. We demonstrate that CLEC5A is homodimeric at the cell surface and binds to dengue virus serotypes 1-4. We used blotting experiments, surface analyses, glycan microarray, and docking studies to investigate the ligand binding potential of CLEC5A with particular respect to dengue virus. This study provides a rational foundation for understanding the dengue virus-macrophage interaction and the role of CLEC5A in dengue virus-induced lethal disease.


Assuntos
Vírus da Dengue/metabolismo , Dengue/metabolismo , Lectinas Tipo C , Macrófagos/metabolismo , Multimerização Proteica , Receptores de Superfície Celular , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cristalografia por Raios X , Dengue/virologia , Células HEK293 , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/metabolismo , Macrófagos/virologia , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Relação Estrutura-Atividade
13.
Commun Biol ; 4(1): 772, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162985

RESUMO

Monoclonal antibodies (mAb) and natural ligands targeting costimulatory tumor necrosis factor receptors (TNFR) exhibit a wide range of agonistic activities and antitumor responses. The mechanisms underlying these differential agonistic activities remain poorly understood. Here, we employ a panel of experimental and clinically-relevant molecules targeting human CD40, 4-1BB and OX40 to examine this issue. Confocal and STORM microscopy reveal that strongly agonistic reagents induce clusters characterized by small area and high receptor density. Using antibody pairs differing only in isotype we show that hIgG2 confers significantly more receptor clustering than hIgG1 across all three receptors, explaining its greater agonistic activity, with receptor clustering shielding the receptor-agonist complex from further molecular access. Nevertheless, discrete receptor clustering patterns are observed with different hIgG2 mAb, with a unique rod-shaped assembly observed with the most agonistic mAb. These findings dispel the notion that larger receptor clusters elicit greater agonism, and instead point to receptor density and subsequent super-structure as key determinants.


Assuntos
Receptores do Fator de Necrose Tumoral/agonistas , Animais , Anticorpos Monoclonais/farmacologia , Afinidade de Anticorpos , Antígenos CD40/agonistas , Antígenos CD40/química , Linhagem Celular , Humanos , Imunoglobulina G/farmacologia , Camundongos , Microscopia Confocal , Receptores OX40/agonistas , Receptores do Fator de Necrose Tumoral/química , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas
14.
Front Cell Dev Biol ; 9: 673446, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34368126

RESUMO

The Jurkat E6.1 clone has been extensively used as a powerful tool for the genetic and biochemical dissection of the TCR signaling pathway. More recently, these cells have been exploited in imaging studies to identify key players in immunological synapse (IS) assembly in superantigen-specific conjugates and to track the dynamics of signaling molecules on glass surfaces coated with activating anti-CD3 antibodies. By comparison, Jurkat cells have been used only scantily for imaging on supported lipid bilayers (SLBs) incorporating laterally mobile TCR and integrin ligands, which allow to study synaptic rearrangements of surface molecules and the fine architecture of the mature IS, likely due to limitations in the assembly of immune synapses with well-defined architecture. Here we have explored whether upregulating the low levels of endogenous LFA-1 expression on Jurkat E6.1 cells through transduction with CD11a- and CD18-encoding lentiviruses can improve IS architecture. We show that, while forced LFA-1 expression did not affect TCR recruitment to the IS, E6.1 LFA-1 high cells assembled better structured synapses, with a tighter distribution of signaling-competent TCRs at the center of the IS. LFA-1 upregulation enhanced protein phosphotyrosine signaling on SLBs but not at the IS formed in conjugates with SEE-pulsed APCs, and led to the constitutive formation of an intracellular phosphotyrosine pool co-localizing with endosomal CD3ζ. This was paralleled by an increase in the levels of p-ZAP-70 and p-Erk both under basal conditions and following activation, and in enhanced Ca2+ mobilization from intracellular stores. The enhancement in early signaling E6.1 LFA-1 high cells did not affect expression of the early activation marker CD69 but led to an increase in IL-2 expression. Our results highlight a new role for LFA-1 in the core architecture of the IS that can be exploited to study the spatiotemporal redistribution of surface receptors on SLBs, thereby extending the potential of E6.1 cells and their derivatives for fine-scale imaging studies.

15.
Neuro Oncol ; 23(7): 1087-1099, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33508126

RESUMO

BACKGROUND: Genome-wide DNA methylation profiling has recently been developed into a tool that allows tumor classification in central nervous system tumors. Extracellular vesicles (EVs) are released by tumor cells and contain high molecular weight DNA, rendering EVs a potential biomarker source to identify tumor subgroups, stratify patients and monitor therapy by liquid biopsy. We investigated whether the DNA in glioblastoma cell-derived EVs reflects genome-wide tumor methylation and mutational profiles and allows noninvasive tumor subtype classification. METHODS: DNA was isolated from EVs secreted by glioblastoma cells as well as from matching cultured cells and tumors. EV-DNA was localized and quantified by direct stochastic optical reconstruction microscopy. Methylation and copy number profiling was performed using 850k arrays. Mutations were identified by targeted gene panel sequencing. Proteins were differentially quantified by mass spectrometric proteomics. RESULTS: Genome-wide methylation profiling of glioblastoma-derived EVs correctly identified the methylation class of the parental cells and original tumors, including the MGMT promoter methylation status. Tumor-specific mutations and copy number variations (CNV) were detected in EV-DNA with high accuracy. Different EV isolation techniques did not affect the methylation profiling and CNV results. DNA was present inside EVs and on the EV surface. Proteome analysis did not allow specific tumor identification or classification but identified tumor-associated proteins that could potentially be useful for enriching tumor-derived circulating EVs from biofluids. CONCLUSIONS: This study provides proof of principle that EV-DNA reflects the genome-wide methylation, CNV, and mutational status of glioblastoma cells and enables their molecular classification.


Assuntos
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , DNA/metabolismo , Variações do Número de Cópias de DNA , Metilação de DNA , Vesículas Extracelulares/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Metilação
16.
Sci Adv ; 7(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33187978

RESUMO

Using AI, we identified baricitinib as having antiviral and anticytokine efficacy. We now show a 71% (95% CI 0.15 to 0.58) mortality benefit in 83 patients with moderate-severe SARS-CoV-2 pneumonia with few drug-induced adverse events, including a large elderly cohort (median age, 81 years). An additional 48 cases with mild-moderate pneumonia recovered uneventfully. Using organotypic 3D cultures of primary human liver cells, we demonstrate that interferon-α2 increases ACE2 expression and SARS-CoV-2 infectivity in parenchymal cells by greater than fivefold. RNA-seq reveals gene response signatures associated with platelet activation, fully inhibited by baricitinib. Using viral load quantifications and superresolution microscopy, we found that baricitinib exerts activity rapidly through the inhibition of host proteins (numb-associated kinases), uniquely among antivirals. This reveals mechanistic actions of a Janus kinase-1/2 inhibitor targeting viral entry, replication, and the cytokine storm and is associated with beneficial outcomes including in severely ill elderly patients, data that incentivize further randomized controlled trials.


Assuntos
Antivirais/farmacologia , Azetidinas/farmacologia , COVID-19/mortalidade , Inibidores Enzimáticos/farmacologia , Janus Quinases/antagonistas & inibidores , Fígado/virologia , Purinas/farmacologia , Pirazóis/farmacologia , SARS-CoV-2/patogenicidade , Sulfonamidas/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19/metabolismo , COVID-19/virologia , Síndrome da Liberação de Citocina , Citocinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Perfilação da Expressão Gênica , Humanos , Interferon alfa-2/metabolismo , Itália , Janus Quinases/metabolismo , Fígado/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Segurança do Paciente , Ativação Plaquetária , Modelos de Riscos Proporcionais , RNA-Seq , Espanha , Internalização do Vírus/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
17.
Wellcome Open Res ; 5: 42, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-36865034

RESUMO

Background: The leukaemia-derived Jurkat E6.1 cell line has been used as a model T cell in the study of many aspects of T cell biology, most notably activation in response to T cell receptor (TCR) engagement. Methods: We present whole-transcriptome RNA-Sequencing data for Jurkat E6.1 cells in the resting state and two hours post-activation via TCR and CD28. We compare early transcriptional responses in the presence and absence of the chemokines CXCL12 and CCL19, and perform a basic comparison between observed transcriptional responses in Jurkat E6.1 cells and those in primary human T cells using publicly deposited data. Results: Jurkat E6.1 cells have many of the hallmarks of standard T cell transcriptional responses to activation, but lack most of the depth of responses in primary cells. Conclusions: These data indicate that Jurkat E6.1 cells hence represent only a highly simplified model of early T cell transcriptional responses.

18.
Front Cell Dev Biol ; 8: 608484, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33537301

RESUMO

A central process in immunity is the activation of T cells through interaction of T cell receptors (TCRs) with agonistic peptide-major histocompatibility complexes (pMHC) on the surface of antigen presenting cells (APCs). TCR-pMHC binding triggers the formation of an extensive contact between the two cells termed the immunological synapse, which acts as a platform for integration of multiple signals determining cellular outcomes, including those from multiple co-stimulatory/inhibitory receptors. Contributors to this include a number of chemokine receptors, notably CXC-chemokine receptor 4 (CXCR4), and other members of the G protein-coupled receptor (GPCR) family. Although best characterized as mediators of ligand-dependent chemotaxis, some chemokine receptors are also recruited to the synapse and contribute to signaling in the absence of ligation. How these and other GPCRs integrate within the dynamic structure of the synapse is unknown, as is how their normally migratory Gαi-coupled signaling is terminated upon recruitment. Here, we report the spatiotemporal organization of several GPCRs, focusing on CXCR4, and the G protein Gαi2 within the synapse of primary human CD4+ T cells on supported lipid bilayers, using standard- and super-resolution fluorescence microscopy. We find that CXCR4 undergoes orchestrated phases of reorganization, culminating in recruitment to the TCR-enriched center. This appears to be dependent on CXCR4 ubiquitination, and does not involve stable interactions with TCR microclusters, as viewed at the nanoscale. Disruption of this process by mutation impairs CXCR4 contributions to cellular activation. Gαi2 undergoes active exclusion from the synapse, partitioning from centrally-accumulated CXCR4. Using a CRISPR-Cas9 knockout screen, we identify several diverse GPCRs with contributions to T cell activation, most significantly the sphingosine-1-phosphate receptor S1PR1, and the oxysterol receptor GPR183. These, and other GPCRs, undergo organization similar to CXCR4; including initial exclusion, centripetal transport, and lack of receptor-TCR interactions. These constitute the first observations of GPCR dynamics within the synapse, and give insights into how these receptors may contribute to T cell activation. The observation of broad GPCR contributions to T cell activation also opens the possibility that modulating GPCR expression in response to cell status or environment may directly regulate responsiveness to pMHC.

19.
Methods Mol Biol ; 1947: 183-197, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30969417

RESUMO

How G protein-coupled receptors are assembled is a matter of considerable interest owing in large part to their remarkable pharmacological importance. For determining receptor stoichiometry, resonance energy transfer-based methods offer considerable advantages insofar as they provide the necessary spatial resolution, and because measurements can be made in situ, relatively easily. This chapter describes three complementary stoichiometric assays that rely on measurements of bioluminescence resonance energy transfer. These quantitative approaches make it possible to identify true protein-protein interactions from non-specific associations that inevitably result from constraining proteins in cellular membranes. In our experience, concordant data obtained in two or more of these assays, benchmarked with suitable controls, strongly predict receptor stoichiometry.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Membrana Celular/metabolismo , Luciferases de Renilla/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Fluorescência , Células HEK293 , Humanos , Ligantes , Ligação Proteica , Conformação Proteica
20.
J Cell Biol ; 217(9): 2983-2985, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30108125

RESUMO

Natural killer cells target antibody-bound cells following engagement of the Fc receptor CD16. Srpan et al. (2018. J. Cell Biol. https://doi.org/10.1083/jcb.201712085) reveal that activation-induced shedding of CD16 leads to more motile behavior, allowing more targets to be engaged and killed in a given time.


Assuntos
Células Matadoras Naturais , Receptores Fc , Sinapses
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa