Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 623
Filtrar
Más filtros

Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
Annu Rev Cell Dev Biol ; 30: 79-109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25103867

RESUMEN

Pathogens use a vast number of strategies to alter host membrane dynamics. Targeting the host membrane machinery is important for the survival and pathogenesis of several extracellular, vacuolar, and cytosolic bacteria. Membrane manipulation promotes bacterial replication while suppressing host responses, allowing the bacterium to thrive in a hostile environment. This review provides a comprehensive summary of various strategies used by both extracellular and intracellular bacteria to hijack host membrane trafficking machinery. We start with mechanisms used by bacteria to alter the plasma membrane, delve into the hijacking of various vesicle trafficking pathways, and conclude by summarizing bacterial adaptation to host immune responses. Understanding bacterial manipulation of host membrane trafficking provides insights into bacterial pathogenesis and uncovers the molecular mechanisms behind various processes within a eukaryotic cell.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Membrana Celular/metabolismo , Células/microbiología , Interacciones Huésped-Patógeno/fisiología , Animales , Autofagia/fisiología , Proteínas Bacterianas/fisiología , Toxinas Bacterianas/farmacología , Transporte Biológico , Permeabilidad de la Membrana Celular , Células/ultraestructura , Citosol/microbiología , Endocitosis/fisiología , Humanos , Lisosomas/fisiología , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Fagosomas/fisiología , Transporte de Proteínas , Vacuolas/microbiología , Vacuolas/fisiología
6.
Nat Rev Mol Cell Biol ; 16(8): 461-72, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26177004

RESUMEN

Autophagy is a conserved catabolic process that degrades cytoplasmic constituents and organelles in the lysosome. Starvation-induced protein degradation is a salient feature of autophagy but recent progress has illuminated how autophagy, during both starvation and nutrient-replete conditions, can mobilize diverse cellular energy and nutrient stores such as lipids, carbohydrates and iron. Processes such as lipophagy, glycophagy and ferritinophagy enable cells to salvage key metabolites to sustain and facilitate core anabolic functions. Here, we discuss the established and emerging roles of autophagy in fuelling biosynthetic capacity and in promoting metabolic and nutrient homeostasis.


Asunto(s)
Autofagia , Metabolismo Energético , Adipogénesis , Animales , Metabolismo de los Hidratos de Carbono , Humanos , Hierro/metabolismo , Fagosomas/fisiología , Transporte de Proteínas , Proteolisis
7.
Cell ; 146(5): 682-95, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21884931

RESUMEN

Genetic inhibition of autophagy induces degenerative changes in mammalian tissues that resemble those associated with aging, and normal and pathological aging are often associated with a reduced autophagic potential. Pharmacological or genetic manipulations that increase life span in model organisms often stimulate autophagy, and its inhibition compromises the longevity-promoting effects of caloric restriction, Sirtuin 1 activation, inhibition of insulin/insulin growth factor signaling, or the administration of rapamycin, resveratrol, or spermidine. Here, we discuss the probable cause and effect relationship between perturbed autophagy and aging, as well as possible molecular mechanisms that may mediate the anti-aging effects of autophagy.


Asunto(s)
Envejecimiento , Autofagia , Mamíferos/fisiología , Animales , Muerte Celular , Homeostasis , Humanos , Mitocondrias/fisiología , Fagosomas/fisiología , Levaduras/citología
8.
Immunity ; 44(6): 1392-405, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27287411

RESUMEN

Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.


Asunto(s)
Colitis/inmunología , Células Epiteliales/inmunología , Enfermedades Inflamatorias del Intestino/genética , Lisosomas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Salmonella/inmunología , Salmonella enterica/inmunología , Salmonella typhimurium/inmunología , Animales , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagosomas/fisiología , Polimorfismo Genético , Receptores Acoplados a Proteínas G/genética , Riesgo
9.
Nat Rev Mol Cell Biol ; 14(12): 759-74, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24201109

RESUMEN

Healthy cells use autophagy as a general 'housekeeping' mechanism and to survive stress, including stress induced by nutrient deprivation. Autophagy is initiated at the isolation membrane (originally termed the phagophore), and the coordinated action of ATG (autophagy-related) proteins results in the expansion of this membrane to form the autophagosome. Although the biogenesis of the isolation membrane and the autophagosome is complex and incompletely understood, insight has been gained into the molecular processes involved in initiating the isolation membrane, the source from which this originates (for example, it was recently proposed that the isolation membrane forms from the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM)) and the role of ATG proteins and the vesicular trafficking machinery in autophagosome formation.


Asunto(s)
Autofagia , Fagosomas/fisiología , Animales , Endocitosis , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/fisiología , Humanos , Membranas Intracelulares/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Mitocondrias/metabolismo , Complejos Multiproteicos/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología , Proteínas de Transporte Vesicular/metabolismo
10.
EMBO J ; 39(2): e102020, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31821587

RESUMEN

For CD8 T lymphocytes to mount responses to cancer and virally-infected cells, dendritic cells must capture antigens present in tissues and display them as peptides bound to MHC-I molecules. This is most often accomplished through a pathway called antigen cross-presentation (XPT). Here, we report that the vesicular trafficking protein Rab39a is needed for optimal cross-presentation by dendritic cells in vitro and cross-priming of CD8 T cells in vivo. Without Rab39a, MHC-I presentation of intraphagosomal peptides is inhibited, indicating that Rab39a converts phagosomes into peptide-loading compartments. In this process, Rab39a promotes the delivery of MHC-I molecules from the endoplasmic reticulum (ER) to phagosomes, and increases the levels of peptide-empty MHC-I conformers that can be loaded with peptide in this compartment. Rab39a also increases the levels of Sec22b and NOX2, previously recognized to participate in cross-presentation, on phagosomes, thereby filling in a missing link into how phagosomes mature into cross-presenting vesicles.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Retículo Endoplásmico/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Fagosomas/fisiología , Proteínas de Unión al GTP rab/fisiología , Animales , Retículo Endoplásmico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis , Transporte de Proteínas
11.
Mol Cell ; 59(4): 522-39, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26295960

RESUMEN

Autophagy constitutes a prominent mechanism through which eukaryotic cells preserve homeostasis in baseline conditions and in response to perturbations of the intracellular or extracellular microenvironment. Autophagic responses can be relatively non-selective or target a specific subcellular compartment. At least in part, this depends on the balance between the availability of autophagic substrates ("offer") and the cellular need of autophagic products or functions for adaptation ("demand"). Irrespective of cargo specificity, adaptive autophagy relies on a panel of sensors that detect potentially dangerous cues and convert them into signals that are ultimately relayed to the autophagic machinery. Here, we summarize the molecular systems through which specific subcellular compartments-including the nucleus, mitochondria, plasma membrane, reticular apparatus, and cytosol-convert homeostatic perturbations into an increased offer of autophagic substrates or an accrued cellular demand for autophagic products or functions.


Asunto(s)
Autofagia , Núcleo Celular/fisiología , Retículo Endoplásmico/fisiología , Mitocondrias/fisiología , Animales , Membrana Celular/fisiología , Humanos , Lisosomas/fisiología , Potencial de la Membrana Mitocondrial , Fagosomas/fisiología
12.
Mol Cell ; 57(2): 219-34, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25578879

RESUMEN

Phosphatidylinositol 3-phosphate (PI(3)P), the product of class III PI3K VPS34, recruits specific autophagic effectors, like WIPI2, during the initial steps of autophagosome biogenesis and thereby regulates canonical autophagy. However, mammalian cells can produce autophagosomes through enigmatic noncanonical VPS34-independent pathways. Here we show that PI(5)P can regulate autophagy via PI(3)P effectors and thereby identify a mechanistic explanation for forms of noncanonical autophagy. PI(5)P synthesis by the phosphatidylinositol 5-kinase PIKfyve was required for autophagosome biogenesis, and it increased levels of PI(5)P, stimulated autophagy, and reduced the levels of autophagic substrates. Inactivation of VPS34 impaired recruitment of WIPI2 and DFCP1 to autophagic precursors, reduced ATG5-ATG12 conjugation, and compromised autophagosome formation. However, these phenotypes were rescued by PI(5)P in VPS34-inactivated cells. These findings provide a mechanistic framework for alternative VPS34-independent autophagy-initiating pathways, like glucose starvation, and unravel a cytoplasmic function for PI(5)P, which previously has been linked predominantly to nuclear roles.


Asunto(s)
Autofagia , Fagosomas/fisiología , Fosfatos de Fosfatidilinositol/fisiología , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/metabolismo , Células HeLa , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
13.
Nature ; 533(7601): 115-9, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27096368

RESUMEN

Defects in clearance of dying cells have been proposed to underlie the pathogenesis of systemic lupus erythematosus (SLE). Mice lacking molecules associated with dying cell clearance develop SLE-like disease, and phagocytes from patients with SLE often display defective clearance and increased inflammatory cytokine production when exposed to dying cells in vitro. Previously, we and others described a form of noncanonical autophagy known as LC3-associated phagocytosis (LAP), in which phagosomes containing engulfed particles, including dying cells, recruit elements of the autophagy pathway to facilitate maturation of phagosomes and digestion of their contents. Genome-wide association studies have identified polymorphisms in the Atg5 (ref. 8) and possibly Atg7 (ref. 9) genes, involved in both canonical autophagy and LAP, as markers of a predisposition for SLE. Here we describe the consequences of defective LAP in vivo. Mice lacking any of several components of the LAP pathway show increased serum levels of inflammatory cytokines and autoantibodies, glomerular immune complex deposition, and evidence of kidney damage. When dying cells are injected into LAP-deficient mice, they are engulfed but not efficiently degraded and trigger acute elevation of pro-inflammatory cytokines but not anti-inflammatory interleukin (IL)-10. Repeated injection of dying cells into LAP-deficient, but not LAP-sufficient, mice accelerated the development of SLE-like disease, including increased serum levels of autoantibodies. By contrast, mice deficient in genes required for canonical autophagy but not LAP do not display defective dying cell clearance, inflammatory cytokine production, or SLE-like disease, and, like wild-type mice, produce IL-10 in response to dying cells. Therefore, defects in LAP, rather than canonical autophagy, can cause SLE-like phenomena, and may contribute to the pathogenesis of SLE.


Asunto(s)
Autofagia , Inflamación/patología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Autoanticuerpos/sangre , Autofagia/genética , Citocinas/biosíntesis , Citocinas/sangre , Inflamación/sangre , Inflamación/genética , Interleucina-10/biosíntesis , Riñón/metabolismo , Riñón/patología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fagocitos/citología , Fagocitos/fisiología , Fagosomas/fisiología
14.
Lab Invest ; 101(11): 1467-1474, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34504306

RESUMEN

The mortality rates among patients who initially survive sepsis are, in part, associated with a high risk of secondary lung infections and respiratory failure. Given that phagolysosomes are important for intracellular killing of pathogenic microbes, we investigated how severe lung infections associated with post-sepsis immunosuppression affect phagolysosome biogenesis. In mice with P. aeruginosa-induced pneumonia, we found a depletion of both phagosomes and lysosomes, as evidenced by decreased amounts of microtubule associated protein light chain 3-II (LC3-II) and lysosomal-associated membrane protein (LAMP1). We also found a loss of transcription factor E3 (TFE3) and transcription factor EB (TFEB), which are important activators for transcription of genes encoding autophagy and lysosomal proteins. These events were associated with increased expression of ZKSCAN3, a repressor for transcription of genes encoding autophagy and lysosomal proteins. Zkscan3-/- mice had increased expression of genes involved in the autophagy-lysosomal pathway along with enhanced killing of P. aeruginosa in the lungs, as compared to wild-type mice. These findings highlight the involvement of ZKSCAN3 in response to severe lung infection, including susceptibility to secondary bacterial infections due to immunosuppression.


Asunto(s)
Fagosomas/fisiología , Neumonía Bacteriana/complicaciones , Infecciones por Pseudomonas/complicaciones , Sepsis/inmunología , Factores de Transcripción/deficiencia , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Tolerancia Inmunológica , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Neumonía Bacteriana/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Sepsis/microbiología
15.
Hum Mol Genet ; 28(20): 3355-3368, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31332443

RESUMEN

MicroRNA-204 (miR-204) is expressed in pulmonary, renal, mammary and eye tissue, and its reduction can result in multiple diseases including cancer. We first generated miR-204-/- mice to study the impact of miR-204 loss on retinal and retinal pigment epithelium (RPE) structure and function. The RPE is fundamentally important for maintaining the health and integrity of the retinal photoreceptors. miR-204-/- eyes evidenced areas of hyper-autofluorescence and defective photoreceptor digestion, along with increased microglia migration to the RPE. Migratory Iba1+ microglial cells were localized to the RPE apical surface where they participated in the phagocytosis of photoreceptor outer segments (POSs) and contributed to a persistent build-up of rhodopsin. These structural, molecular and cellular outcomes were accompanied by decreased light-evoked electrical responses from the retina and RPE. In parallel experiments, we suppressed miR-204 expression in primary cultures of human RPE using anti-miR-204. In vitro suppression of miR-204 in human RPE similarly showed abnormal POS clearance and altered expression of autophagy-related proteins and Rab22a, a regulator of endosome maturation. Together, these in vitro and in vivo experiments suggest that the normally high levels of miR-204 in RPE can mitigate disease onset by preventing generation of oxidative stress and inflammation originating from intracellular accumulation of undigested photoreactive POS lipids. More generally, these results implicate RPE miR-204-mediated regulation of autophagy and endolysosomal interaction as a critical determinant of normal RPE/retina structure and function.


Asunto(s)
MicroARNs/metabolismo , Fagocitosis/fisiología , Fagosomas/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Electrofisiología , Femenino , Citometría de Flujo , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Fagocitosis/genética , Fagosomas/fisiología , Retina/fisiología , Epitelio Pigmentado de la Retina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nat Immunol ; 10(9): 943-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19692995

RESUMEN

The progression of tuberculosis from a latent, subclinical infection to active disease that culminates in the transmission of infectious bacilli is determined locally at the level of the granuloma. This progression takes place even in the face of a robust immune response that, although it contains infection, is unable to eliminate the bacterium. The factors or environmental conditions that influence this progression remain to be determined. Recent advances have indicated that pathogen-induced dysregulation of host lipid synthesis and sequestration serves a critical role in this transition. The foamy macrophage seems to be a key participant in both sustaining persistent bacteria and contributing to the tissue pathology that leads to cavitation and the release of infectious bacilli.


Asunto(s)
Células Espumosas/fisiología , Granuloma/etiología , Tuberculosis/inmunología , Animales , Progresión de la Enfermedad , Granuloma/inmunología , Granuloma/patología , Humanos , Isocitratoliasa/fisiología , Lípidos/biosíntesis , Lipoproteínas LDL/metabolismo , Fagosomas/fisiología , Tuberculosis/patología , Factor de Necrosis Tumoral alfa/fisiología
17.
Immunity ; 36(6): 947-58, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22658522

RESUMEN

The adaptor protein Bcl10 is a critically important mediator of T cell receptor (TCR)-to-NF-κB signaling. Bcl10 degradation is a poorly understood biological phenomenon suggested to reduce TCR activation of NF-κB. Here we have shown that TCR engagement triggers the degradation of Bcl10 in primary effector T cells but not in naive T cells. TCR engagement promoted K63 polyubiquitination of Bcl10, causing Bcl10 association with the autophagy adaptor p62. Paradoxically, p62 binding was required for both Bcl10 signaling to NF-κB and gradual degradation of Bcl10 by autophagy. Bcl10 autophagy was highly selective, as shown by the fact that it spared Malt1, a direct Bcl10 binding partner. Blockade of Bcl10 autophagy enhanced TCR activation of NF-κB. Together, these data demonstrate that selective autophagy of Bcl10 is a pathway-intrinsic homeostatic mechanism that modulates TCR signaling to NF-κB in effector T cells. This homeostatic process may protect T cells from adverse consequences of unrestrained NF-κB activation, such as cellular senescence.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Autofagia/fisiología , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Relacionadas con la Autofagia , Proteína 10 de la LLC-Linfoma de Células B , Caspasas/fisiología , Diferenciación Celular , Citosol/inmunología , Citosol/ultraestructura , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiología , Homeostasis , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias/fisiología , Fagosomas/fisiología , Fagosomas/ultraestructura , Mapeo de Interacción de Proteínas , Proteína Sequestosoma-1 , Transducción de Señal/genética , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/ultraestructura , Células Th2/inmunología , Células Th2/ultraestructura , Enzimas Ubiquitina-Conjugadoras/fisiología
18.
J Toxicol Environ Health A ; 84(4): 152-172, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33148135

RESUMEN

Inhalation of particles results in pulmonary inflammation; however, treatments are currently lacking. Docosahexaenoic acid (DHA) is an omega-3 polyunsaturated fatty acid shown to exhibit anti-inflammatory capabilities. The impact of DHA on particle-induced inflammation is unclear; therefore, the aim of this study was to examine the hypothesis that DHA downregulates macrophage inflammatory responses by altering phagolysosomal membrane permeability (LMP) and shifting macrophage phenotype. Isolated Balb/c alveolar macrophages (AM) were polarized into M1, M2a, M2b, or M2c phenotypes in vitro, treated with DHA, and exposed to a multi-walled carbon nanotube (MWNCT) or crystalline silica (SiO2). Results showed minimal cytotoxicity, robust effects for silica particle uptake, and LMP differences between phenotypes. Docosahexaenoic acid prevented these effects to the greatest extent in M2c phenotype. To determine if DHA affected inflammation similarly in vivo, Balb/c mice were placed on a control or 1% DHA diet for 3 weeks, instilled with the same particles, and assessed 24 hr following instillation. Data demonstrated that in contrast to in vitro findings, DHA increased pulmonary inflammation and LMP. These results suggest that pulmonary responses in vivo may not necessarily be predicted from single-cell responses in vitro.


Asunto(s)
Antiinflamatorios/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Lisosomas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Material Particulado/toxicidad , Fagosomas/efectos de los fármacos , Animales , Permeabilidad de la Membrana Celular/fisiología , Regulación hacia Abajo , Femenino , Lisosomas/fisiología , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Fagosomas/fisiología
19.
Fish Physiol Biochem ; 47(4): 869-880, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33770301

RESUMEN

The trials of finding non-conventional and alternative aquafeed ingredients are increasing. In this sense, this study evaluated the influence of coconut oil on the growth, feed utilization, immune, and antioxidative responses of Nile tilapia. Five test diets were formulated by mixing coconut oil with the other ingredients at 0, 1, 2, 3, and 4% of the total ration and presented for tilapia for 60 successive days. The final weight, SGR, weight gain (WG), and feed intake were superior in fish delivered 2% of coconut oil (P < 0.05). Concurrently, fish that received 2% coconut oil had lower FCR and higher PER than fish of the control and 4% groups (P < 0.05). Higher lipase activity was observed in fish of 2% and 3% levels than the remaining groups (P < 0.05). Besides, the amylase and protease activities of fish in 1%, 2%, and 3% groups were higher than the 0% level (P < 0.05). The total blood cholesterol, RBCs, and PCV showed higher values in Nile tilapia fed 2% and 3% coconut oil (P < 0.05). The lysozyme and phagocytic activities were higher in fish fed 2% and 3% levels than the control (P < 0.05), while the phagocytic index in 2% and 3% levels was higher than 0% and 4% levels. Furthermore, SOD and CAT were higher in fish fed 1%, 2%, and 3% than fish fed 0% and 4% levels while GSH was higher in fish of 1%, 2%, and 3% than fish fed 0% level (P < 0.05). However, the MDA level was markedly lower in fish fed 25, 3%, and 4% coconut oil than the 0% level (P < 0.05). The intestine's histological structure in all groups appeared normal, forming of intestinal villi projecting from the intestinal wall. Also, the structure of the hepatopancreas had a normal architecture in all groups. To sum up, the inclusion of coconut oil at 2 to 3% is recommended as a replacer for fish oil in Nile tilapia diets.


Asunto(s)
Cíclidos , Aceite de Coco/farmacología , Suplementos Dietéticos , Amilasas/metabolismo , Animales , Antioxidantes , Acuicultura/métodos , Cíclidos/anatomía & histología , Cíclidos/crecimiento & desarrollo , Cíclidos/inmunología , Cíclidos/metabolismo , Hepatopáncreas/anatomía & histología , Intestinos/anatomía & histología , Intestinos/enzimología , Lipasa/metabolismo , Hígado/anatomía & histología , Péptido Hidrolasas/metabolismo , Fagosomas/efectos de los fármacos , Fagosomas/fisiología
20.
Trends Immunol ; 38(6): 407-422, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28416446

RESUMEN

Recognition of microbial pathogens and dead cells and their phagocytic uptake by specialized immune cells are essential to maintain host homeostasis. Phagosomes undergo fusion and fission events with endosomal and lysosomal compartments, a process called 'phagosome maturation', which leads to the degradation of the phagosomal content. However, many phagocytic cells also act as antigen-presenting cells and must balance degradation and peptide preservation. Emerging evidence indicates that receptor engagement by phagosomal cargo, as well as inflammatory mediators and cellular activation affect many aspects of phagosome maturation. Unsurprisingly, pathogens have developed strategies to hijack this machinery, thereby interfering with host immunity. Here, we highlight progress in this field, summarize findings on the impact of immune signals, and discuss consequences for pathogen elimination.


Asunto(s)
Inflamación/metabolismo , Fagocitos/inmunología , Fagosomas/fisiología , Animales , Presentación de Antígeno , Diferenciación Celular , Endosomas/metabolismo , Humanos , Inmunidad Innata , Lisosomas/metabolismo , Fusión de Membrana , Proteolisis , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA