Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.271
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(15): 3981-3997.e22, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34157301

RESUMEN

A fraction of mature T cells can be activated by peripheral self-antigens, potentially eliciting host autoimmunity. We investigated homeostatic control of self-activated T cells within unperturbed tissue environments by combining high-resolution multiplexed and volumetric imaging with computational modeling. In lymph nodes, self-activated T cells produced interleukin (IL)-2, which enhanced local regulatory T cell (Treg) proliferation and inhibitory functionality. The resulting micro-domains reciprocally constrained inputs required for damaging effector responses, including CD28 co-stimulation and IL-2 signaling, constituting a negative feedback circuit. Due to these local constraints, self-activated T cells underwent transient clonal expansion, followed by rapid death ("pruning"). Computational simulations and experimental manipulations revealed the feedback machinery's quantitative limits: modest reductions in Treg micro-domain density or functionality produced non-linear breakdowns in control, enabling self-activated T cells to subvert pruning. This fine-tuned, paracrine feedback process not only enforces immune homeostasis but also establishes a sharp boundary between autoimmune and host-protective T cell responses.


Asunto(s)
Retroalimentación Fisiológica , Homeostasis/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Interleucina-2/metabolismo , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Modelos Inmunológicos , Comunicación Paracrina , Transducción de Señal
2.
Cell ; 176(5): 1040-1053.e17, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30712872

RESUMEN

Sphingomyelin and cholesterol are essential lipids that are enriched in plasma membranes of animal cells, where they interact to regulate membrane properties and many intracellular signaling processes. Despite intense study, the interaction between these lipids in membranes is not well understood. Here, structural and biochemical analyses of ostreolysin A (OlyA), a protein that binds to membranes only when they contain both sphingomyelin and cholesterol, reveal that sphingomyelin adopts two distinct conformations in membranes when cholesterol is present. One conformation, bound by OlyA, is induced by stoichiometric, exothermic interactions with cholesterol, properties that are consistent with sphingomyelin/cholesterol complexes. In its second conformation, sphingomyelin is free from cholesterol and does not bind OlyA. A point mutation abolishes OlyA's ability to discriminate between these two conformations. In cells, levels of sphingomyelin/cholesterol complexes are held constant over a wide range of plasma membrane cholesterol concentrations, enabling precise regulation of the chemical activity of cholesterol.


Asunto(s)
Membrana Celular/ultraestructura , Esfingomielinas/metabolismo , Esfingomielinas/fisiología , Animales , Línea Celular , Membrana Celular/metabolismo , Colesterol/metabolismo , Colesterol/fisiología , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/ultraestructura , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/ultraestructura , Humanos , Microdominios de Membrana/metabolismo , Conformación Molecular
3.
Cell ; 177(4): 806-819, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31051105

RESUMEN

Over the last several decades, an impressive array of advanced microscopic and analytical tools, such as single-particle tracking and nanoscopic fluorescence correlation spectroscopy, has been applied to characterize the lateral organization and mobility of components in the plasma membrane. Such analysis can tell researchers about the local dynamic composition and structure of membranes and is important for predicting the outcome of membrane-based reactions. However, owing to the unresolved complexity of the membrane and the structures peripheral to it, identification of the detailed molecular origin of the interactions that regulate the organization and mobility of the membrane has not proceeded quickly. This Perspective presents an overview of how cell-surface structure may give rise to the types of lateral mobility that are observed and some potentially fruitful future directions to elucidate the architecture of these structures in more molecular detail.


Asunto(s)
Membrana Celular/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Membrana Celular/fisiología , Membrana Dobles de Lípidos/química , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/química , Proteínas de la Membrana/fisiología
4.
Cell ; 177(7): 1738-1756.e23, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31104842

RESUMEN

Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are a major class of lipid-anchored plasma membrane proteins. GPI-APs form nanoclusters generated by cortical acto-myosin activity. While our understanding of the physical principles governing this process is emerging, the molecular machinery and functional relevance of GPI-AP nanoclustering are unknown. Here, we first show that a membrane receptor signaling pathway directs nanocluster formation. Arg-Gly-Asp motif-containing ligands bound to the ß1-integrin receptor activate src and focal adhesion kinases, resulting in RhoA signaling. This cascade triggers actin-nucleation via specific formins, which, along with myosin activity, drive the nanoclustering of membrane proteins with actin-binding domains. Concurrently, talin-mediated activation of the mechano-transducer vinculin is required for the coupling of the acto-myosin machinery to inner-leaflet lipids, thereby generating GPI-AP nanoclusters. Second, we show that these nanoclusters are functional; disruption of their formation either in GPI-anchor remodeling mutants or in vinculin mutants impairs cell spreading and migration, hallmarks of integrin function.


Asunto(s)
Integrina beta1/metabolismo , Mecanotransducción Celular , Microdominios de Membrana/metabolismo , Secuencias de Aminoácidos , Animales , Células CHO , Cricetulus , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrina beta1/genética , Microdominios de Membrana/genética , Vinculina/genética , Vinculina/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
5.
Immunity ; 57(6): 1378-1393.e14, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38749447

RESUMEN

Tumors weakly infiltrated by T lymphocytes poorly respond to immunotherapy. We aimed to unveil malignancy-associated programs regulating T cell entrance, arrest, and activation in the tumor environment. Differential expression of cell adhesion and tissue architecture programs, particularly the presence of the membrane tetraspanin claudin (CLDN)18 as a signature gene, demarcated immune-infiltrated from immune-depleted mouse pancreatic tumors. In human pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer, CLDN18 expression positively correlated with more differentiated histology and favorable prognosis. CLDN18 on the cell surface promoted accrual of cytotoxic T lymphocytes (CTLs), facilitating direct CTL contacts with tumor cells by driving the mobilization of the adhesion protein ALCAM to the lipid rafts of the tumor cell membrane through actin. This process favored the formation of robust immunological synapses (ISs) between CTLs and CLDN18-positive cancer cells, resulting in increased T cell activation. Our data reveal an immune role for CLDN18 in orchestrating T cell infiltration and shaping the tumor immune contexture.


Asunto(s)
Carcinoma Ductal Pancreático , Claudinas , Activación de Linfocitos , Neoplasias Pancreáticas , Linfocitos T Citotóxicos , Animales , Humanos , Ratones , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Claudinas/metabolismo , Claudinas/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Sinapsis Inmunológicas/metabolismo , Sinapsis Inmunológicas/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Microdominios de Membrana/metabolismo , Microdominios de Membrana/inmunología , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología
6.
Cell ; 171(6): 1354-1367.e20, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29103614

RESUMEN

A number of bacterial cell processes are confined functional membrane microdomains (FMMs), structurally and functionally similar to lipid rafts of eukaryotic cells. How bacteria organize these intricate platforms and what their biological significance is remain important questions. Using the pathogen methicillin-resistant Staphylococcus aureus (MRSA), we show here that membrane-carotenoid interaction with the scaffold protein flotillin leads to FMM formation, which can be visualized using super-resolution array tomography. These membrane platforms accumulate multimeric protein complexes, for which flotillin facilitates efficient oligomerization. One of these proteins is PBP2a, responsible for penicillin resistance in MRSA. Flotillin mutants are defective in PBP2a oligomerization. Perturbation of FMM assembly using available drugs interferes with PBP2a oligomerization and disables MRSA penicillin resistance in vitro and in vivo, resulting in MRSA infections that are susceptible to penicillin treatment. Our study demonstrates that bacteria possess sophisticated cell organization programs and defines alternative therapies to fight multidrug-resistant pathogens using conventional antibiotics.


Asunto(s)
Microdominios de Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/fisiología , Infecciones Estafilocócicas/microbiología , Animales , Proteínas Bacterianas/metabolismo , Carotenoides/metabolismo , Membrana Celular/metabolismo , Femenino , Microdominios de Membrana/química , Proteínas de la Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Proteínas de Unión a las Penicilinas/metabolismo , Xantófilas/metabolismo
7.
Cell ; 164(4): 722-34, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26853472

RESUMEN

Diverse cellular processes are driven by motor proteins that are recruited to and generate force on lipid membranes. Surprisingly little is known about how membranes control the force from motors and how this may impact specific cellular functions. Here, we show that dynein motors physically cluster into microdomains on the membrane of a phagosome as it matures inside cells. Such geometrical reorganization allows many dyneins within a cluster to generate cooperative force on a single microtubule. This results in rapid directed transport of the phagosome toward microtubule minus ends, likely promoting phagolysosome fusion and pathogen degradation. We show that lipophosphoglycan, the major molecule implicated in immune evasion of Leishmania donovani, inhibits phagosome motion by disrupting the clustering and therefore the cooperative force generation of dynein. These findings appear relevant to several pathogens that prevent phagosome-lysosome fusion by targeting lipid microdomains on phagosomes.


Asunto(s)
Leishmania donovani/citología , Leishmania donovani/metabolismo , Lisosomas/metabolismo , Fagosomas/metabolismo , Animales , Transporte Biológico , Línea Celular , Dictyostelium/citología , Dineínas/metabolismo , Glicoesfingolípidos/metabolismo , Microdominios de Membrana/metabolismo , Ratones
8.
Nature ; 632(8025): 664-671, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39048819

RESUMEN

Biological membranes are partitioned into functional zones termed membrane microdomains, which contain specific lipids and proteins1-3. The composition and organization of membrane microdomains remain controversial because few techniques are available that allow the visualization of lipids in situ without disrupting their native behaviour3,4. The yeast eisosome, composed of the BAR-domain proteins Pil1 and Lsp1 (hereafter, Pil1/Lsp1), scaffolds a membrane compartment that senses and responds to mechanical stress by flattening and releasing sequestered factors5-9. Here we isolated near-native eisosomes as helical tubules made up of a lattice of Pil1/Lsp1 bound to plasma membrane lipids, and solved their structures by helical reconstruction. Our structures reveal a striking organization of membrane lipids, and, using in vitro reconstitutions and molecular dynamics simulations, we confirmed the positioning of individual PI(4,5)P2, phosphatidylserine and sterol molecules sequestered beneath the Pil1/Lsp1 coat. Three-dimensional variability analysis of the native-source eisosomes revealed a dynamic stretching of the Pil1/Lsp1 lattice that affects the sequestration of these lipids. Collectively, our results support a mechanism in which stretching of the Pil1/Lsp1 lattice liberates lipids that would otherwise be anchored by the Pil1/Lsp1 coat, and thus provide mechanistic insight into how eisosome BAR-domain proteins create a mechanosensitive membrane microdomain.


Asunto(s)
Microscopía por Crioelectrón , Microdominios de Membrana , Simulación de Dinámica Molecular , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestructura , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Microdominios de Membrana/ultraestructura , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Modelos Moleculares , Esteroles/química , Esteroles/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Dominios Proteicos , Estrés Mecánico , Fosfoproteínas
9.
Nat Rev Mol Cell Biol ; 18(6): 361-374, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28356571

RESUMEN

Cellular plasma membranes are laterally heterogeneous, featuring a variety of distinct subcompartments that differ in their biophysical properties and composition. A large number of studies have focused on understanding the basis for this heterogeneity and its physiological relevance. The membrane raft hypothesis formalized a physicochemical principle for a subtype of such lateral membrane heterogeneity, in which the preferential associations between cholesterol and saturated lipids drive the formation of relatively packed (or ordered) membrane domains that selectively recruit certain lipids and proteins. Recent studies have yielded new insights into this mechanism and its relevance in vivo, owing primarily to the development of improved biochemical and biophysical technologies.


Asunto(s)
Membrana Celular/química , Membrana Celular/metabolismo , Lípidos de la Membrana/química , Lípidos de la Membrana/metabolismo , Animales , Humanos , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo
10.
Nat Immunol ; 17(10): 1150-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548432

RESUMEN

The innate immune system needs to distinguish between harmful and innocuous stimuli to adapt its activation to the level of threat. How Drosophila mounts differential immune responses to dead and live Gram-negative bacteria using the single peptidoglycan receptor PGRP-LC is unknown. Here we describe rPGRP-LC, an alternative splice variant of PGRP-LC that selectively dampens immune response activation in response to dead bacteria. rPGRP-LC-deficient flies cannot resolve immune activation after Gram-negative infection and die prematurely. The alternative exon in the encoding gene, here called rPGRP-LC, encodes an adaptor module that targets rPGRP-LC to membrane microdomains and interacts with the negative regulator Pirk and the ubiquitin ligase DIAP2. We find that rPGRP-LC-mediated resolution of an efficient immune response requires degradation of activating and regulatory receptors via endosomal ESCRT sorting. We propose that rPGRP-LC selectively responds to peptidoglycans from dead bacteria to tailor the immune response to the level of threat.


Asunto(s)
Proteínas Portadoras/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/metabolismo , Infecciones por Bacterias Gramnegativas/inmunología , Microdominios de Membrana/metabolismo , Pectobacterium carotovorum/inmunología , Isoformas de ARN/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Exones/genética , Técnicas de Inactivación de Genes , Inmunidad/genética , Inmunomodulación , Proteínas Inhibidoras de la Apoptosis/metabolismo , Unión Proteica , Señales de Clasificación de Proteína/genética , Proteolisis , Isoformas de ARN/genética , Relación Estructura-Actividad
11.
Mol Cell ; 73(5): 971-984.e5, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661983

RESUMEN

Both the timing and kinetics of neurotransmitter release depend on the positioning of clustered Ca2+ channels in active zones to docked synaptic vesicles on presynaptic plasma membranes. However, how active zones form is not known. Here, we show that RIM and RIM-BP, via specific multivalent bindings, form dynamic and condensed assemblies through liquid-liquid phase separation. Voltage-gated Ca2+ channels (VGCCs), via C-terminal-tail-mediated direct binding to both RIM and RIM-BP, can be enriched to the RIM and RIM-BP condensates. We further show that RIM and RIM-BP, together with VGCCs, form dense clusters on the supported lipid membrane bilayers via phase separation. Therefore, RIMs and RIM-BPs are plausible organizers of active zones, and the formation of RIM and RIM-BP condensates may cluster VGCCs into nano- or microdomains and position the clustered Ca2+ channels with Ca2+ sensors on docked vesicles for efficient and precise synaptic transmissions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Canales de Calcio Tipo N/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Terminales Presinápticos/metabolismo , Membranas Sinápticas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Sitios de Unión , Canales de Calcio Tipo N/genética , Proteínas de Unión al GTP/genética , Proteínas Intrínsecamente Desordenadas/genética , Proteínas Intrínsecamente Desordenadas/metabolismo , Cinética , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Ratas , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Solubilidad , Membranas Sinápticas/genética , Transmisión Sináptica
12.
J Cell Sci ; 137(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38668720

RESUMEN

Amyloid ß (Aß) is a central contributor to neuronal damage and cognitive impairment in Alzheimer's disease (AD). Aß disrupts AMPA receptor-mediated synaptic plasticity, a key factor in early AD progression. Numerous studies propose that Aß oligomers hinder synaptic plasticity, particularly long-term potentiation (LTP), by disrupting GluA1 (encoded by GRIA1) function, although the precise mechanism remains unclear. In this study, we demonstrate that Aß mediates the accumulation of GM1 ganglioside in lipid raft domains of cultured cells, and GluA1 exhibits preferential localization in lipid rafts via direct binding to GM1. Aß enhances the raft localization of GluA1 by increasing GM1 in these areas. Additionally, chemical LTP stimulation induces lipid raft-dependent GluA1 internalization in Aß-treated neurons, resulting in reduced cell surface and postsynaptic expression of GluA1. Consistent with this, disrupting lipid rafts and GluA1 localization in rafts rescues Aß-mediated suppression of hippocampal LTP. These findings unveil a novel functional deficit in GluA1 trafficking induced by Aß, providing new insights into the mechanism underlying AD-associated cognitive dysfunction.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Hipocampo , Potenciación a Largo Plazo , Microdominios de Membrana , Receptores AMPA , Péptidos beta-Amiloides/metabolismo , Receptores AMPA/metabolismo , Microdominios de Membrana/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Hipocampo/metabolismo , Gangliósido G(M1)/metabolismo , Humanos , Neuronas/metabolismo , Ratas , Ratones , Transporte de Proteínas
13.
Cell ; 146(4): 593-606, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21854984

RESUMEN

The mechanisms that govern receptor coalescence into functional clusters--often a critical step in their stimulation by ligand--are poorly understood. We used single-molecule tracking to investigate the dynamics of CD36, a clustering-responsive receptor that mediates oxidized LDL uptake by macrophages. We found that CD36 motion in the membrane was spatially structured by the cortical cytoskeleton. A subpopulation of receptors diffused within linear confinement regions whose unique geometry simultaneously facilitated freedom of movement along one axis while increasing the effective receptor density. Co-confinement within troughs enhanced the probability of collisions between unligated receptors and promoted their clustering. Cytoskeleton perturbations that inhibited diffusion in linear confinement regions reduced receptor clustering in the absence of ligand and, following ligand addition, suppressed CD36-mediated signaling and internalization. These observations demonstrate a role for the cytoskeleton in controlling signal transduction by structuring receptor diffusion within membrane regions that increase their collision frequency.


Asunto(s)
Antígenos CD36/metabolismo , Citoesqueleto/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Actomiosina/metabolismo , Línea Celular , Células Cultivadas , Humanos , Macrófagos/citología , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Microtúbulos/metabolismo , Pinzas Ópticas
14.
Nature ; 588(7838): 491-497, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33149299

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) has previously been identified as an endosomal protein that blocks viral infection1-3. Here we studied clinical cohorts of patients with B cell leukaemia and lymphoma, and identified IFITM3 as a strong predictor of poor outcome. In normal resting B cells, IFITM3 was minimally expressed and mainly localized in endosomes. However, engagement of the B cell receptor (BCR) induced both expression of IFITM3 and phosphorylation of this protein at Tyr20, which resulted in the accumulation of IFITM3 at the cell surface. In B cell leukaemia, oncogenic kinases phosphorylate IFITM3 at Tyr20, which causes constitutive localization of this protein at the plasma membrane. In a mouse model, Ifitm3-/- naive B cells developed in normal numbers; however, the formation of germinal centres and the production of antigen-specific antibodies were compromised. Oncogenes that induce the development of leukaemia and lymphoma did not transform Ifitm3-/- B cells. Conversely, the phosphomimetic IFITM3(Y20E) mutant induced oncogenic PI3K signalling and initiated the transformation of premalignant B cells. Mechanistic experiments revealed that IFITM3 functions as a PIP3 scaffold and central amplifier of PI3K signalling. The amplification of PI3K signals depends on IFITM3 using two lysine residues (Lys83 and Lys104) in its conserved intracellular loop as a scaffold for the accumulation of PIP3. In Ifitm3-/- B cells, lipid rafts were depleted of PIP3, which resulted in the defective expression of over 60 lipid-raft-associated surface receptors, and impaired BCR signalling and cellular adhesion. We conclude that the phosphorylation of IFITM3 that occurs after B cells encounter antigen induces a dynamic switch from antiviral effector functions in endosomes to a PI3K amplification loop at the cell surface. IFITM3-dependent amplification of PI3K signalling, which in part acts downstream of the BCR, is critical for the rapid expansion of B cells with high affinity to antigen. In addition, multiple oncogenes depend on IFITM3 to assemble PIP3-dependent signalling complexes and amplify PI3K signalling for malignant transformation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Animales , Antígenos CD19/metabolismo , Linfocitos B/enzimología , Linfocitos B/inmunología , Linfocitos B/patología , Transformación Celular Neoplásica , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/patología , Humanos , Integrinas/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Modelos Moleculares , Fosforilación , Receptores de Antígenos de Linfocitos B/metabolismo
15.
Annu Rev Cell Dev Biol ; 28: 215-50, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22905956

RESUMEN

The recent rapid accumulation of knowledge on the dynamics and structure of the plasma membrane has prompted major modifications of the textbook fluid-mosaic model. However, because the new data have been obtained in a variety of research contexts using various biological paradigms, the impact of the critical conceptual modifications on biomedical research and development has been limited. In this review, we try to synthesize our current biological, chemical, and physical knowledge about the plasma membrane to provide new fundamental organizing principles of this structure that underlie every molecular mechanism that realizes its functions. Special attention is paid to signal transduction function and the dynamic aspect of the organizing principles. We propose that the cooperative action of the hierarchical three-tiered mesoscale (2-300 nm) domains--actin-membrane-skeleton induced compartments (40-300 nm), raft domains (2-20 nm), and dynamic protein complex domains (3-10 nm)--is critical for membrane function and distinguishes the plasma membrane from a classical Singer-Nicolson-type model.


Asunto(s)
Microdominios de Membrana/metabolismo , Modelos Biológicos , Transducción de Señal , Animales , Membrana Celular/metabolismo , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Humanos , Microdominios de Membrana/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Estructura Cuaternaria de Proteína
16.
Proc Natl Acad Sci U S A ; 120(48): e2312603120, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-37983503

RESUMEN

Plasma membrane heterogeneity is a key biophysical regulatory principle of membrane protein dynamics, which further influences downstream signal transduction. Although extensive biophysical and cell biology studies have proven membrane heterogeneity is essential to cell fate, the direct link between membrane heterogeneity regulation to cellular function remains unclear. Heterogeneous structures on plasma membranes, such as lipid rafts, are transiently assembled, thus hard to study via regular techniques. Indeed, it is nearly impossible to perturb membrane heterogeneity without changing plasma membrane compositions. In this study, we developed a high-spatial resolved DNA-origami-based nanoheater system with specific lipid heterogeneity targeting to manipulate the local lipid environmental temperature under near-infrared (NIR) laser illumination. Our results showed that the targeted heating of the local lipid environment influences the membrane thermodynamic properties, which further triggers an integrin-associated cell migration change. Therefore, the nanoheater system was further applied as an optimized therapeutic agent for wound healing. Our strategy provides a powerful tool to dynamically manipulate membrane heterogeneity and has the potential to explore cellular function through changes in plasma membrane biophysical properties.


Asunto(s)
Calor , Microdominios de Membrana , Membrana Celular/metabolismo , Microdominios de Membrana/metabolismo , Transducción de Señal , Movimiento Celular , Lípidos/análisis
17.
Annu Rev Biochem ; 79: 803-33, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20196649

RESUMEN

Although viruses are simple in structure and composition, their interactions with host cells are complex. Merely to gain entry, animal viruses make use of a repertoire of cellular processes that involve hundreds of cellular proteins. Although some viruses have the capacity to penetrate into the cytosol directly through the plasma membrane, most depend on endocytic uptake, vesicular transport through the cytoplasm, and delivery to endosomes and other intracellular organelles. The internalization may involve clathrin-mediated endocytosis (CME), macropinocytosis, caveolar/lipid raft-mediated endocytosis, or a variety of other still poorly characterized mechanisms. This review focuses on the cell biology of virus entry and the different strategies and endocytic mechanisms used by animal viruses.


Asunto(s)
Endocitosis , Internalización del Virus , Animales , Caveolas/metabolismo , Clatrina/metabolismo , Microdominios de Membrana/metabolismo , Fagocitosis , Pinocitosis
18.
J Biol Chem ; 300(4): 107154, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38479603

RESUMEN

Styrene-maleic acid (SMA) and similar amphiphilic copolymers are known to cut biological membranes into lipid nanoparticles/nanodiscs containing membrane proteins apparently in their relatively native membrane lipid environment. Our previous work demonstrated that membrane raft microdomains resist such disintegration by SMA. The use of SMA in studying membrane proteins is limited by its heterogeneity and the inability to prepare defined derivatives. In the present paper, we demonstrate that some amphiphilic peptides structurally mimicking SMA also similarly disintegrate cell membranes. In contrast to the previously used copolymers, the simple peptides are structurally homogeneous. We found that their membrane-disintegrating activity increases with their length (reaching optimum at 24 amino acids) and requires a basic primary structure, that is, (XXD)n, where X represents a hydrophobic amino acid (optimally phenylalanine), D aspartic acid, and n is the number of repeats of these triplets. These peptides may provide opportunities for various well-defined potentially useful modifications in the study of membrane protein biochemistry. Our present results confirm a specific character of membrane raft microdomains.


Asunto(s)
Proteínas de la Membrana , Péptidos , Animales , Humanos , Membrana Celular/metabolismo , Membrana Celular/química , Maleatos/química , Microdominios de Membrana/metabolismo , Microdominios de Membrana/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Péptidos/química , Poliestirenos/química , Línea Celular
19.
Nat Immunol ; 14(1): 82-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23202272

RESUMEN

Phosphorylation of the T cell antigen receptor (TCR) by the tyrosine kinase Lck is an essential step in the activation of T cells. Because Lck is constitutively active, spatial organization may regulate TCR signaling. Here we found that Lck distributions on the molecular level were controlled by the conformational states of Lck, with the open, active conformation inducing clustering and the closed, inactive conformation preventing clustering. In contrast, association with lipid domains and protein networks were not sufficient or necessary for Lck clustering. Conformation-driven Lck clustering was highly dynamic, so that TCR triggering resulted in Lck clusters that contained phosphorylated TCRs but excluded the phosphatase CD45. Our data suggest that Lck conformational states represent an intrinsic mechanism for the intermolecular organization of early T cell signaling.


Asunto(s)
Regulación Alostérica , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Conformación Proteica , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Humanos , Células Jurkat , Activación de Linfocitos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Proteínas Mutantes/genética , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal , Relación Estructura-Actividad , Transgenes/genética
20.
Nat Chem Biol ; 19(6): 750-758, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36997644

RESUMEN

Plasma membrane heterogeneity has been tied to a litany of cellular functions and is often explained by analogy to membrane phase separation; however, models based on phase separation alone fall short of describing the rich organization available within cell membranes. Here we present comprehensive experimental evidence motivating an updated model of plasma membrane heterogeneity in which membrane domains assemble in response to protein scaffolds. Quantitative super-resolution nanoscopy measurements in live B lymphocytes detect membrane domains that emerge upon clustering B cell receptors (BCRs). These domains enrich and retain membrane proteins based on their preference for the liquid-ordered phase. Unlike phase-separated membranes that consist of binary phases with defined compositions, membrane composition at BCR clusters is modulated through the protein constituents in clusters and the composition of the membrane overall. This tunable domain structure is detected through the variable sorting of membrane probes and impacts the magnitude of BCR activation.


Asunto(s)
Microdominios de Membrana , Proteínas de la Membrana , Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA