Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 5.074
Filtrar
Más filtros

Publication year range
1.
Cell ; 184(24): 5950-5969.e22, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34741801

RESUMEN

The biogenesis of mammalian autophagosomes remains to be fully defined. Here, we used cellular and in vitro membrane fusion analyses to show that autophagosomes are formed from a hitherto unappreciated hybrid membrane compartment. The autophagic precursors emerge through fusion of FIP200 vesicles, derived from the cis-Golgi, with endosomally derived ATG16L1 membranes to generate a hybrid pre-autophagosomal structure, HyPAS. A previously unrecognized apparatus defined here controls HyPAS biogenesis and mammalian autophagosomal precursor membranes. HyPAS can be modulated by pharmacological agents whereas its formation is inhibited upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or by expression of SARS-CoV-2 nsp6. These findings reveal the origin of mammalian autophagosomal membranes, which emerge via convergence of secretory and endosomal pathways, and show that this process is targeted by microbial factors such as coronaviral membrane-modulating proteins.


Asunto(s)
Autofagosomas/virología , COVID-19/virología , Autofagia , COVID-19/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Endosomas/fisiología , Endosomas/virología , Aparato de Golgi/fisiología , Células HEK293 , Células HeLa , Humanos , Fusión de Membrana , Microscopía Confocal , Fagosomas/metabolismo , Fagosomas/virología , Proteínas Qa-SNARE/biosíntesis , Receptores sigma/biosíntesis , SARS-CoV-2 , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/biosíntesis , Sinaptotagminas/biosíntesis , Receptor Sigma-1
2.
Cell ; 180(1): 135-149.e14, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31883797

RESUMEN

Autophagy is a conserved catabolic homeostasis process central for cellular and organismal health. During autophagy, small single-membrane phagophores rapidly expand into large double-membrane autophagosomes to encapsulate diverse cargoes for degradation. It is thought that autophagic membranes are mainly derived from preformed organelle membranes. Instead, here we delineate a pathway that expands the phagophore membrane by localized phospholipid synthesis. Specifically, we find that the conserved acyl-CoA synthetase Faa1 accumulates on nucleated phagophores and locally activates fatty acids (FAs) required for phagophore elongation and autophagy. Strikingly, using isotopic FA tracing, we directly show that Faa1 channels activated FAs into the synthesis of phospholipids and promotes their assembly into autophagic membranes. Indeed, the first committed steps of de novo phospholipid synthesis at the ER, which forms stable contacts with nascent autophagosomes, are essential for autophagy. Together, our work illuminates how cells spatially tune synthesis and flux of phospholipids for autophagosome biogenesis during autophagy.


Asunto(s)
Autofagia/fisiología , Ácidos Grasos/metabolismo , Fagosomas/metabolismo , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Membrana Celular/metabolismo , Coenzima A Ligasas/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo de los Lípidos , Proteínas de la Membrana/metabolismo , Fagosomas/fisiología , Fosfolípidos/biosíntesis , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Cell ; 179(1): 74-89.e10, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31495570

RESUMEN

During neural tube closure and spinal cord development, many cells die in both the central and peripheral nervous systems (CNS and PNS, respectively). However, myeloid-derived professional phagocytes have not yet colonized the trunk region during early neurogenesis. How apoptotic cells are removed from this region during these stages remains largely unknown. Using live imaging in zebrafish, we demonstrate that neural crest cells (NCCs) respond rapidly to dying cells and phagocytose cellular debris around the neural tube. Additionally, NCCs have the ability to enter the CNS through motor exit point transition zones and clear debris in the spinal cord. Surprisingly, NCCs phagocytosis mechanistically resembles macrophage phagocytosis and their recruitment toward cellular debris is mediated by interleukin-1ß. Taken together, our results reveal a role for NCCs in phagocytosis of debris in the developing nervous system before the presence of professional phagocytes.


Asunto(s)
Movimiento Celular/fisiología , Cresta Neural/fisiología , Neurogénesis/fisiología , Sistema Nervioso Periférico/crecimiento & desarrollo , Fagocitosis/fisiología , Médula Espinal/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Diferenciación Celular/fisiología , Interleucina-1beta/metabolismo , Fagocitos/fisiología , Fagosomas/fisiología , Pez Cebra/embriología
4.
Nat Immunol ; 22(2): 140-153, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33349708

RESUMEN

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.


Asunto(s)
Presentación de Antígeno , Reactividad Cruzada , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Fagosomas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Linfocitos T/metabolismo , Animales , Muerte Celular , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células HEK293 , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Lectinas Tipo C/genética , Ligandos , Ratones , NADPH Oxidasas/metabolismo , Fagosomas/genética , Fagosomas/inmunología , Fosforilación , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/genética , Receptores Mitogénicos/genética , Transducción de Señal , Quinasa Syk/metabolismo , Linfocitos T/inmunología
5.
Nat Rev Mol Cell Biol ; 22(11): 733-750, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34302147

RESUMEN

Autophagy is a versatile degradation system for maintaining cellular homeostasis whereby cytosolic materials are sequestered in a double-membrane autophagosome and subsequently delivered to lysosomes, where they are broken down. In multicellular organisms, newly formed autophagosomes undergo a process called 'maturation', in which they fuse with vesicles originating from endolysosomal compartments, including early/late endosomes and lysosomes, to form amphisomes, which eventually become degradative autolysosomes. This fusion process requires the concerted actions of multiple regulators of membrane dynamics, including SNAREs, tethering proteins and RAB GTPases, and also transport of autophagosomes and late endosomes/lysosomes towards each other. Multiple mechanisms modulate autophagosome maturation, including post-translational modification of key components, spatial distribution of phosphoinositide lipid species on membranes, RAB protein dynamics, and biogenesis and function of lysosomes. Nutrient status and various stresses integrate into the autophagosome maturation machinery to coordinate the progression of autophagic flux. Impaired autophagosome maturation is linked to the pathogenesis of various human diseases, including neurodegenerative disorders, cancer and myopathies. Furthermore, invading pathogens exploit various strategies to block autophagosome maturation, thus evading destruction and even subverting autophagic vacuoles (autophagosomes, amphisomes and autolysosomes) for survival, growth and/or release. Here, we discuss the recent progress in our understanding of the machinery and regulation of autophagosome maturation, the relevance of these mechanisms to human pathophysiology and how they are harnessed by pathogens for their benefit. We also provide perspectives on targeting autophagosome maturation therapeutically.


Asunto(s)
Autofagosomas/genética , Autofagia/genética , Enfermedades Neurodegenerativas/genética , Vesículas Transportadoras/genética , Endosomas/genética , Humanos , Lisosomas/genética , Enfermedades Neurodegenerativas/patología , Fagosomas/genética , Procesamiento Proteico-Postraduccional/genética , Proteínas SNARE/genética , Proteínas de Unión al GTP rab/genética
6.
Cell ; 175(2): 429-441.e16, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30245008

RESUMEN

Targeting autophagy in cancer cells and in the tumor microenvironment are current goals of cancer therapy. However, components of canonical autophagy play roles in other biological processes, adding complexity to this goal. One such alternative function of autophagy proteins is LC3-associated phagocytosis (LAP), which functions in phagosome maturation and subsequent signaling events. Here, we show that impairment of LAP in the myeloid compartment, rather than canonical autophagy, induces control of tumor growth by tumor-associated macrophages (TAM) upon phagocytosis of dying tumor cells. Single-cell RNA sequencing (RNA-seq) analysis revealed that defects in LAP induce pro-inflammatory gene expression and trigger STING-mediated type I interferon responses in TAM. We found that the anti-tumor effects of LAP impairment require tumor-infiltrating T cells, dependent upon STING and the type I interferon response. Therefore, autophagy proteins in the myeloid cells of the tumor microenvironment contribute to immune suppression of T lymphocytes by effecting LAP.


Asunto(s)
Tolerancia Inmunológica/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Fagocitosis/fisiología , Animales , Autofagia/inmunología , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Tolerancia Inmunológica/inmunología , Macrófagos , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Células Mieloides/metabolismo , Fagosomas/fisiología , Linfocitos T/metabolismo , Microambiente Tumoral/fisiología
7.
Cell ; 175(2): 442-457.e23, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290143

RESUMEN

Antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) critically contribute to the efficacy of anti-tumor therapeutic antibodies. We report here an unexpected finding that macrophages after ADCP inhibit NK cell-mediated ADCC and T cell-mediated cytotoxicity in breast cancers and lymphomas. Mechanistically, AIM2 is recruited to the phagosomes by FcγR signaling following ADCP and activated by sensing the phagocytosed tumor DNAs through the disrupted phagosomal membrane, which subsequently upregulates PD-L1 and IDO and causes immunosuppression. Combined treatment with anti-HER2 antibody and inhibitors of PD-L1 and IDO enhances anti-tumor immunity and anti-HER2 therapeutic efficacy in mouse models. Furthermore, neoadjuvant trastuzumab therapy significantly upregulates PD-L1 and IDO in the tumor-associated macrophages (TAMs) of HER2+ breast cancer patients, correlating with poor trastuzumab response. Collectively, our findings unveil a deleterious role of ADCP macrophages in cancer immunosuppression and suggest that therapeutic antibody plus immune checkpoint blockade may provide synergistic effects in cancer treatment.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Citofagocitosis/inmunología , Macrófagos/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/fisiología , Antígeno B7-H1/genética , Antígeno B7-H1/fisiología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citofagocitosis/fisiología , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia , Células Asesinas Naturales/fisiología , Linfoma/inmunología , Macrófagos/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fagocitosis/inmunología , Fagocitosis/fisiología , Fagosomas/fisiología , Receptores de IgG/inmunología
8.
Annu Rev Biochem ; 86: 225-244, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28301741

RESUMEN

Autophagy is the process of cellular self-eating by a double-membrane organelle, the autophagosome. A range of signaling processes converge on two protein complexes to initiate autophagy: the ULK1 (unc51-like autophagy activating kinase 1) protein kinase complex and the PI3KC3-C1 (class III phosphatidylinositol 3-kinase complex I) lipid kinase complex. Some 90% of the mass of these large protein complexes consists of noncatalytic domains and subunits, and the ULK1 complex has essential noncatalytic activities. Structural studies of these complexes have shed increasing light on the regulation of their catalytic and noncatalytic activities in autophagy initiation. The autophagosome is thought to nucleate from vesicles containing the integral membrane protein Atg9 (autophagy-related 9), COPII (coat protein complex II) vesicles, and possibly other sources. In the wake of reconstitution and super-resolution imaging studies, we are beginning to understand how the ULK1 and PI3KC3-C1 complexes might coordinate the nucleation and fusion of Atg9 and COPII vesicles at the start of autophagosome biogenesis.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/química , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/ultraestructura , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Fosfatidilinositol 3-Quinasas Clase III/química , Fosfatidilinositol 3-Quinasas Clase III/genética , Células Eucariotas/metabolismo , Células Eucariotas/ultraestructura , Expresión Génica , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Fagosomas/ultraestructura , Fosfatidilinositol 3-Quinasa/química , Fosfatidilinositol 3-Quinasa/genética , Unión Proteica , Multimerización de Proteína , Transducción de Señal
9.
Immunity ; 56(8): 1727-1742.e6, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37379835

RESUMEN

STING (stimulator of interferon genes) exerts protective cellular responses to viral infection via induction of interferon production and autophagy. Here, we report the role of STING in modulating the immune responses toward fungal infection. Upon Candida albicans stimulation, STING transited alongside the endoplasmic reticulum (ER) to the phagosomes. In phagosomes, STING directly bound with Src via the N-terminal 18 amino acids of STING, and this binding prevented Src from recruiting and phosphorylating Syk. Consistently, Syk-associated signaling and production of pro-inflammatory cytokines and chemokines were increased in mouse BMDCs (bone-marrow-derived dendritic cells) lacking STING with fungal treatment. STING deficiency improved anti-fungal immunity in systemic C. albicans infection. Importantly, administration of the N-terminal 18-aa (amino acid) peptide of STING improved host outcomes in disseminated fungal infection. Overall, our study identifies a previously unrecognized function of STING in negatively regulating anti-fungal immune responses and offers a potential therapeutic strategy for controlling C. albicans infection.


Asunto(s)
Nucleótidos , Transducción de Señal , Animales , Ratones , Citocinas/metabolismo , Inmunidad Innata , Interferones/metabolismo , Nucleótidos/metabolismo , Fagosomas/metabolismo , Fagosomas/microbiología
10.
Nat Rev Mol Cell Biol ; 21(1): 25-42, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31705132

RESUMEN

Cellular membranes can form two principally different involutions, which either exclude or contain cytosol. The 'classical' budding reactions, such as those occurring during endocytosis or formation of exocytic vesicles, involve proteins that assemble on the cytosol-excluding face of the bud neck. Inverse membrane involution occurs in a wide range of cellular processes, supporting cytokinesis, endosome maturation, autophagy, membrane repair and many other processes. Such inverse membrane remodelling is mediated by a heteromultimeric protein machinery known as endosomal sorting complex required for transport (ESCRT). ESCRT proteins assemble on the cytosolic (or nucleoplasmic) face of the neck of the forming involution and cooperate with the ATPase VPS4 to drive membrane scission or sealing. Here, we review similarities and differences of various ESCRT-dependent processes, with special emphasis on mechanisms of ESCRT recruitment.


Asunto(s)
Membrana Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Neuronas/citología , Membrana Nuclear/metabolismo , Replicación Viral/fisiología , Animales , Citocinesis , Endosomas/metabolismo , Exosomas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Neuronas/metabolismo , Fagosomas/metabolismo , Transporte de Proteínas , Espastina/metabolismo
11.
Cell ; 171(2): 331-345.e22, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942921

RESUMEN

Clearance of apoptotic cells (ACs) by phagocytes (efferocytosis) prevents post-apoptotic necrosis and dampens inflammation. Defective efferocytosis drives important diseases, including atherosclerosis. For efficient efferocytosis, phagocytes must be able to internalize multiple ACs. We show here that uptake of multiple ACs by macrophages requires dynamin-related protein 1 (Drp1)-mediated mitochondrial fission, which is triggered by AC uptake. When mitochondrial fission is disabled, AC-induced increase in cytosolic calcium is blunted owing to mitochondrial calcium sequestration, and calcium-dependent phagosome formation around secondarily encountered ACs is impaired. These defects can be corrected by silencing the mitochondrial calcium uniporter (MCU). Mice lacking myeloid Drp1 showed defective efferocytosis and its pathologic consequences in the thymus after dexamethasone treatment and in advanced atherosclerotic lesions in fat-fed Ldlr-/- mice. Thus, mitochondrial fission in response to AC uptake is a critical process that enables macrophages to clear multiple ACs and to avoid the pathologic consequences of defective efferocytosis in vivo.


Asunto(s)
Macrófagos/citología , Dinámicas Mitocondriales , Animales , Apoptosis , Humanos , Macrófagos/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Células Mieloides/metabolismo , Fagocitos/metabolismo , Fagosomas/metabolismo
12.
Annu Rev Biochem ; 85: 685-713, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26865532

RESUMEN

Autophagy is a conserved intracellular pathway that delivers cytoplasmic contents to lysosomes for degradation via double-membrane autophagosomes. Autophagy substrates include organelles such as mitochondria, aggregate-prone proteins that cause neurodegeneration and various pathogens. Thus, this pathway appears to be relevant to the pathogenesis of diverse diseases, and its modulation may have therapeutic value. Here, we focus on the cell and molecular biology of mammalian autophagy and review the key proteins that regulate the process by discussing their roles and how these may be modulated by posttranslational modifications. We consider the membrane-trafficking events that impact autophagy and the questions relating to the sources of autophagosome membrane(s). Finally, we discuss data from structural studies and some of the insights these have provided.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas SNARE/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/genética , Fosfatidilinositol 3-Quinasas Clase III/genética , Citoesqueleto/química , Citoesqueleto/metabolismo , Endocitosis , Humanos , Lisosomas/metabolismo , Mamíferos , Modelos Moleculares , Fagosomas/metabolismo , Proteínas SNARE/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab/genética
13.
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
14.
Cell ; 162(5): 1101-12, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26317472

RESUMEN

Potassium is the most abundant ion to face both plasma and organelle membranes. Extensive research over the past seven decades has characterized how K(+) permeates the plasma membrane to control fundamental processes such as secretion, neuronal communication, and heartbeat. However, how K(+) permeates organelles such as lysosomes and endosomes is unknown. Here, we directly recorded organelle K(+) conductance and discovered a major K(+)-selective channel KEL on endosomes and lysosomes. KEL is formed by TMEM175, a protein with unknown function. Unlike any of the ∼80 plasma membrane K(+) channels, TMEM175 has two repeats of 6-transmembrane-spanning segments and has no GYG K(+) channel sequence signature-containing, pore-forming P loop. Lysosomes lacking TMEM175 exhibit no K(+) conductance, have a markedly depolarized ΔΨ and little sensitivity to changes in [K(+)], and have compromised luminal pH stability and abnormal fusion with autophagosomes during autophagy. Thus, TMEM175 comprises a K(+) channel that underlies the molecular mechanism of lysosomal K(+) permeability.


Asunto(s)
Endosomas/metabolismo , Lisosomas/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Membranas Intracelulares/metabolismo , Ratones , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Fagosomas/metabolismo , Canales de Potasio/química , Canales de Potasio/genética , Alineación de Secuencia
15.
Nature ; 630(8017): 736-743, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38839956

RESUMEN

Phagocytosis is the process by which myeloid phagocytes bind to and internalize potentially dangerous microorganisms1. During phagocytosis, innate immune receptors and associated signalling proteins are localized to the maturing phagosome compartment, forming an immune information processing hub brimming with microorganism-sensing features2-8. Here we developed proximity labelling of phagosomal contents (PhagoPL) to identify proteins localizing to phagosomes containing model yeast and bacteria. By comparing the protein composition of phagosomes containing evolutionarily and biochemically distinct microorganisms, we unexpectedly identified programmed death-ligand 1 (PD-L1) as a protein that specifically enriches in phagosomes containing yeast. We found that PD-L1 directly binds to yeast upon processing in phagosomes. By surface display library screening, we identified the ribosomal protein Rpl20b as a fungal protein ligand for PD-L1. Using an auxin-inducible depletion system, we found that detection of Rpl20b by macrophages cross-regulates production of distinct cytokines including interleukin-10 (IL-10) induced by the activation of other innate immune receptors. Thus, this study establishes PhagoPL as a useful approach to quantifying the collection of proteins enriched in phagosomes during host-microorganism interactions, exemplified by identifying PD-L1 as a receptor that binds to fungi.


Asunto(s)
Antígeno B7-H1 , Proteínas Fúngicas , Fagosomas , Proteínas Ribosómicas , Saccharomyces cerevisiae , Animales , Femenino , Humanos , Masculino , Ratones , Antígeno B7-H1/metabolismo , Escherichia coli/metabolismo , Proteínas Fúngicas/metabolismo , Interacciones Microbiota-Huesped , Inmunidad Innata , Interleucina-10/metabolismo , Ligandos , Macrófagos/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Ratones Endogámicos BALB C , Fagocitosis , Fagosomas/química , Fagosomas/metabolismo , Fagosomas/microbiología , Unión Proteica , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Staphylococcus aureus/metabolismo
16.
Nature ; 628(8007): 408-415, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38480883

RESUMEN

During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20-60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches-precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)-on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.


Asunto(s)
Eferocitosis , Macrófagos , ARN Polimerasa II , Elongación de la Transcripción Genética , Animales , Humanos , Masculino , Ratones , Apoptosis , Citoesqueleto/metabolismo , Proteína 3 de la Respuesta de Crecimiento Precoz/deficiencia , Proteína 3 de la Respuesta de Crecimiento Precoz/genética , Eferocitosis/genética , Concentración de Iones de Hidrógeno , Macrófagos/inmunología , Macrófagos/metabolismo , Neuronas/metabolismo , Fagosomas/metabolismo , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Pez Cebra/embriología , Pez Cebra/genética , Factores de Tiempo
17.
Cell ; 157(2): 300-311, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725401

RESUMEN

Autophagy is directed by numerous distinct autophagy-related (Atg) proteins. These transmit starvation-induced signals to lipids and regulatory proteins and assemble a double-membrane autophagosome sequestering bulk cytoplasm and/or selected cargos destined for degradation upon autophagosome fusion with a vacuole or lysosome. This Review discusses the structural mechanisms by which Atg proteins sense membrane curvature, mediate a PI(3)P-signaling cascade, and utilize autophagy-specific ubiquitin-like protein cascades to tether proteins to autophagosomal membranes. Recent elucidation of molecular interactions enabling vesicle nucleation, elongation, and cargo recruitment provides insights into how dynamic protein-protein and protein-membrane interactions may dictate size, shape, and contents of autophagosomes.


Asunto(s)
Autofagia , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Fagosomas/metabolismo
18.
Cell ; 158(3): 506-21, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083866

RESUMEN

Adaptation of the endoplasmic reticulum (ER) pathway for MHC class I (MHC-I) presentation in dendritic cells enables cross-presentation of peptides derived from phagocytosed microbes, infected cells, or tumor cells to CD8 T cells. How these peptides intersect with MHC-I molecules remains poorly understood. Here, we show that MHC-I selectively accumulate within phagosomes carrying microbial components, which engage Toll-like receptor (TLR) signaling. Although cross-presentation requires Sec22b-mediated phagosomal recruitment of the peptide loading complex from the ER-Golgi intermediate compartment (ERGIC), this step is independent of TLR signaling and does not deliver MHC-I. Instead, MHC-I are recruited from an endosomal recycling compartment (ERC), which is marked by Rab11a, VAMP3/cellubrevin, and VAMP8/endobrevin and holds large reserves of MHC-I. While Rab11a activity stocks ERC stores with MHC-I, MyD88-dependent TLR signals drive IκB-kinase (IKK)2-mediated phosphorylation of phagosome-associated SNAP23. Phospho-SNAP23 stabilizes SNARE complexes orchestrating ERC-phagosome fusion, enrichment of phagosomes with ERC-derived MHC-I, and subsequent cross-presentation during infection.


Asunto(s)
Presentación de Antígeno , Endosomas/metabolismo , Fagosomas/metabolismo , Receptores Toll-Like/metabolismo , Animales , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Tejido Linfoide , Ratones , Ovalbúmina/inmunología , Fagocitosis , Fosforilación , Transporte de Proteínas , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Receptores Toll-Like/inmunología , Proteínas de Unión al GTP rab/metabolismo
19.
Nat Immunol ; 17(2): 196-203, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26692173

RESUMEN

Canonically, immunoglobulin E (IgE) mediates allergic immune responses by triggering mast cells and basophils to release histamine and type 2 helper cytokines. Here we found that in human systemic lupus erythematosus (SLE), IgE antibodies specific for double-stranded DNA (dsDNA) activated plasmacytoid dendritic cells (pDCs), a type of cell of the immune system linked to viral defense, which led to the secretion of substantial amounts of interferon-α (IFN-α). The concentration of dsDNA-specific IgE found in patient serum correlated with disease severity and greatly potentiated pDC function by triggering phagocytosis via the high-affinity FcɛRI receptor for IgE, followed by Toll-like receptor 9 (TLR9)-mediated sensing of DNA in phagosomes. Our findings expand the known pathogenic mechanisms of IgE-mediated inflammation beyond those found in allergy and demonstrate that IgE can trigger interferon responses capable of exacerbating self-destructive autoimmune responses.


Asunto(s)
Autoanticuerpos/inmunología , Autoinmunidad , Inmunoglobulina E/inmunología , Interferones/metabolismo , Anticuerpos Antinucleares/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Masculino , Fagocitosis/inmunología , Fagosomas/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptor Toll-Like 9/metabolismo
20.
Nat Rev Mol Cell Biol ; 22(3): 161, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495650
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda