Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Elife ; 122024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38507462

RESUMEN

The trimeric SARS-CoV-2 Spike protein mediates viral attachment facilitating cell entry. Most COVID-19 vaccines direct mammalian cells to express the Spike protein or deliver it directly via inoculation to engender a protective immune response. The trafficking and cellular tropism of the Spike protein in vivo and its impact on immune cells remains incompletely elucidated. In this study, we inoculated mice intranasally, intravenously, and subcutaneously with fluorescently labeled recombinant SARS-CoV-2 Spike protein. Using flow cytometry and imaging techniques, we analyzed its localization, immune cell tropism, and acute functional impact. Intranasal administration led to rapid lung alveolar macrophage uptake, pulmonary vascular leakage, and neutrophil recruitment and damage. When injected near the inguinal lymph node medullary, but not subcapsular macrophages, captured the protein, while scrotal injection recruited and fragmented neutrophils. Widespread endothelial and liver Kupffer cell uptake followed intravenous administration. Human peripheral blood cells B cells, neutrophils, monocytes, and myeloid dendritic cells all efficiently bound Spike protein. Exposure to the Spike protein enhanced neutrophil NETosis and augmented human macrophage TNF-α (tumor necrosis factor-α) and IL-6 production. Human and murine immune cells employed C-type lectin receptors and Siglecs to help capture the Spike protein. This study highlights the potential toxicity of the SARS-CoV-2 Spike protein for mammalian cells and illustrates the central role for alveolar macrophage in pathogenic protein uptake.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Humanos , Ratones , Animales , Glicoproteína de la Espiga del Coronavirus/metabolismo , Macrófagos Alveolares , SARS-CoV-2/metabolismo , Vacunas contra la COVID-19 , Infiltración Neutrófila , Factor de Necrosis Tumoral alfa , Mamíferos/metabolismo
2.
Elife ; 112022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35404237

RESUMEN

B-cell activation and immune synapse (IS) formation with membrane-bound antigens are actin-dependent processes that scale positively with the strength of antigen-induced signals. Importantly, ligating the B-cell integrin, LFA-1, with ICAM-1 promotes IS formation when antigen is limiting. Whether the actin cytoskeleton plays a specific role in integrin-dependent IS formation is unknown. Here, we show using super-resolution imaging of mouse primary B cells that LFA-1:ICAM-1 interactions promote the formation of an actomyosin network that dominates the B-cell IS. This network is created by the formin mDia1, organized into concentric, contractile arcs by myosin 2A, and flows inward at the same rate as B-cell receptor (BCR):antigen clusters. Consistently, individual BCR microclusters are swept inward by individual actomyosin arcs. Under conditions where integrin is required for synapse formation, inhibiting myosin impairs synapse formation, as evidenced by reduced antigen centralization, diminished BCR signaling, and defective signaling protein distribution at the synapse. Together, these results argue that a contractile actomyosin arc network plays a key role in the mechanism by which LFA-1 co-stimulation promotes B-cell activation and IS formation.


The immune system has the ability to recognize a vast array of infections and trigger rapid responses. This defense mechanism is mediated in part by B cells which make antibodies that can neutralize or destroy specific disease-causing agents. When pathogens (such as bacteria or viruses) invade the body, a specialized immune cell called an 'antigen presenting cell' holds it in place and presents it to the B cell to examine. Receptors on the surface of the B cell then bind to the infectious agent and launch the B cell into action, triggering the antibody response needed to remove the pathogen. This process relies on B cells and antigen presenting cells making a close connection called an immune synapse, which has a bulls-eye pattern with the receptor in the middle surrounded by sticky proteins called adhesion molecules. A network of actin filaments coating the inside of the B cell are responsible for arranging the proteins into this bulls-eye shape. Once fully formed, the synapse initiates the production of antibodies and helps B cells to make stronger versions of these defensive proteins. So far, most studies have focused on the role the receptor plays in B cell activation. However, when there are only small amounts of the pathogen available, these receptors bind to the antigen presenting cell very weakly. When this happens, adhesion molecules have been shown to step in and promote the formation of the mature synapse needed for B cell activation. But it is not fully understood how adhesion molecules do this. To investigate, Wang et al. looked at mouse B cells using super resolution microscopes. This revealed that when B cells receive signals through both their receptors and their adhesion molecules, they rearrange their actin into a circular structure composed of arc shapes. Motors on the actin arcs then contract the structure inwards, pushing the B cell receptors into the classic bullseye pattern. This only happened when adhesion molecules were present and signals through the B cell receptors were weak. These findings suggest that adhesion molecules help form immune synapses and activate B cells by modifying the actin network so it can drive the re-patterning of receptor proteins. B cells are responsible for the long-term immunity provided by vaccines. Thus, it is possible that the findings of Wang et al. could be harnessed to create vaccines that trigger a stronger antibody response.


Asunto(s)
Actomiosina , Linfocitos B , Sinapsis Inmunológicas , Antígeno-1 Asociado a Función de Linfocito , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Animales , Linfocitos B/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Miosinas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo
3.
Autophagy ; 18(1): 204-222, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34313548

RESUMEN

CD38 is a cell surface receptor capable of generating calcium-mobilizing second messengers. It has been implicated in host defense and cancer biology, but signaling mechanisms downstream of CD38 remain unclear. Mutations in LRRK2 (leucine-rich repeat kinase 2) are the most common genetic cause of Parkinson disease; it is also a risk factor for Crohn disease, leprosy, and certain types of cancers. The pathogenesis of these diseases involves inflammation and macroautophagy/autophagy, processes both CD38 and LRRK2 are implicated in. Here, we mechanistically and functionally link CD38 and LRRK2 as upstream activators of TFEB (transcription factor EB), a host defense transcription factor and the master transcriptional regulator of the autophagy/lysosome machinery. In B-lymphocytes and macrophages, we show that CD38 and LRRK2 exist in a complex on the plasma membrane. Ligation of CD38 with the monoclonal antibody clone 90 results in internalization of the CD38-LRRK2 complex and its targeting to the endolysosomal system. This generates an NAADP-dependent calcium signal, which requires LRRK2 kinase activity, and results in the downstream activation of TFEB. lrrk2 KO macrophages accordingly have TFEB activation defects following CD38 or LPS stimulation and fail to switch to glycolytic metabolism after LPS treatment. In overexpression models, the pathogenic LRRK2G2019S mutant promotes hyperactivation of TFEB even in the absence of CD38, both by stabilizing TFEB and promoting its nuclear translocation via aberrant calcium signaling. In sum, we have identified a physiological CD38-LRRK2-TFEB signaling axis in immune cells. The common pathogenic mutant, LRRK2G2019S, appears to hijack this pathway.Abbreviations:ADPR: ADP-ribose; AMPK: AMP-activated protein kinase; BMDM: bone marrow-derived macrophage; cADPR: cyclic-ADP-ribose; COR: C-terminal of ROC; CTSD: cathepsin D; ECAR: extracellular acidification rate; EDTA: ethylenediaminetetraacetic acid; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GPN: Gly-Phe ß-naphthylamide; GSK3B/GSK3ß: glycogen synthase kinase 3 beta; GTP: guanosine triphosphate; KD: knockdown; LAMP1: lysosomal-associated membrane protein 1; LRR: leucine rich repeat; LRRK2: leucine rich repeat kinase 2; mAb: monoclonal antibody; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MAPK/ERK: mitogen-activated protein kinase; MCOLN1: mucolipin 1; MFI: mean fluorescence intensity; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; NAADP: nicotinic acid adenine dinucleotide phosphate; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; PD: Parkinson disease; PPP3CB: protein phosphatase 3, catalytic subunit, beta isoform; q-RT-PCR: quantitative reverse transcription polymerase chain reaction; ROC: Ras of complex; siRNA: small interfering RNA; SQSTM1/p62: sequestome 1; TFEB: transcription factor EB; TPCN: two pore channel; TRPM2: transient receptor potential cation channel, subfamily M, member 2; ZKSCAN3: zinc finger with KRAB and SCAN domains 3.


Asunto(s)
Autofagia , Enfermedad de Parkinson , Adenosina Difosfato Ribosa/metabolismo , Anticuerpos Monoclonales , Autofagia/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Calcio/metabolismo , Humanos , Leucina/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Lipopolisacáridos/metabolismo , Lisosomas/metabolismo , NADP/análogos & derivados , NADP/metabolismo , Enfermedad de Parkinson/metabolismo , Factores de Transcripción
4.
Front Immunol ; 12: 679856, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34135907

RESUMEN

Neutrophil trafficking, homeostatic and pathogen elicited, depends upon chemoattractant receptors triggering heterotrimeric G-protein Gαißγ signaling, whose magnitude and kinetics are governed by RGS protein/Gαi interactions. RGS proteins typically limit Gαi signaling by reducing the duration that Gαi subunits remain GTP bound and able to activate downstream effectors. Yet how in totality RGS proteins shape neutrophil chemoattractant receptor activated responses remains unclear. Here, we show that C57Bl/6 mouse neutrophils containing a genomic knock-in of a mutation that disables all RGS protein-Gαi2 interactions (G184S) cannot properly balance chemoattractant receptor signaling, nor appropriately respond to inflammatory insults. Mutant neutrophils accumulate in mouse bone marrow, spleen, lung, and liver; despite neutropenia and an intrinsic inability to properly mobilize from the bone marrow. In vitro they rapidly adhere to ICAM-1 coated plates, but in vivo they poorly adhere to blood vessel endothelium. Those few neutrophils that cross blood vessels and enter tissues migrate haphazardly. Following Concanavalin-A administration fragmented G184S neutrophils accumulate in liver sinusoids leading to thrombo-inflammation and perivasculitis. Thus, neutrophil Gαi2/RGS protein interactions both limit and facilitate Gαi2 signaling thereby promoting normal neutrophil trafficking, aging, and clearance.


Asunto(s)
Senescencia Celular , Quimiotaxis de Leucocito , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Transducción de Señal , Animales , Trasplante de Médula Ósea , Senescencia Celular/genética , Senescencia Celular/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Humanos , Inmunofenotipificación , Masculino , Ratones , Neutropenia/etiología , Neutrófilos/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/metabolismo
5.
J Immunol ; 205(8): 2255-2264, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32929041

RESUMEN

The cytosolic pattern recognition receptor NLRP3 senses host-derived danger signals and certain microbe-derived products in both humans and rodents. NLRP3 activation assembles an inflammasome complex that contains the adapter proteins ASC and caspase-1, whose activation triggers the maturation and release of the proinflammatory cytokines IL-1ß and IL-18. S5 phosphorylation of NLRP3 prevents its oligomerization and activation, whereas dephosphorylation of this residue by the phosphatase PP2A allows NLRP3 activation. However, the protein kinase that mediates NLRP3 S5 phosphorylation is unknown. In this study, we show that AKT associates with NLRP3 and phosphorylates it on S5, limiting NLRP3 oligomerization. This phosphorylation event also stabilizes NLRP3 by reducing its ubiquitination on lysine 496, which inhibits its proteasome-mediated degradation by the E3 ligase Trim31. Pharmacologic manipulation of AKT kinase activity reciprocally modulates NLRP3 inflammasome-mediated IL-1ß production. Inhibition of AKT reduced IL-1ß production following the i.p. injection of LPS into mice. We propose that AKT, Trim31, and PP2A together modulate NLRP3 protein levels and the tendency to oligomerize, thereby setting a tightly regulated threshold for NLRP3 activation.


Asunto(s)
Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Animales , Caspasa 1/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Ratones , Fosforilación/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Proteolisis , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Ubiquitinación/inmunología
6.
Cell Death Discov ; 5: 151, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31839993

RESUMEN

Apoptosis is a form of programmed cell death in multicellular organisms. Bcl-2 prevents apoptosis and promotes cellular survival by neutralizing BH3 domain-containing proteins, which directly activate the pore-forming proteins BAX and BAK. However, Bcl-2 is not known to regulate other cell death effectors such as gasdermin D (GSDMD) or mixed lineage kinase domain-like (MLKL), whose activation causes pyroptosis and necroptosis, respectively. Here, we identify a BH3-like domain in both GSDMD and MLKL that mediates an interaction with B-cell lymphoma 2 (Bcl-2). The presence of Bcl-2 reduced GSDMD cleavage at D275 by caspase-1, 4 or 5, and enhanced the GSDMD cleavage at D87. The GSDMD D87 cleavage inactivates the pyroptotic execution program. The presence of Bcl-2 also limited RIP3 mediated phosphorylation of MLKL, which reduced MLKL oligomerization and tempered the induction of necroptosis. Our observations suggest that the presence of Bcl-2 limits the induction of three forms of cell death apoptosis, pyroptosis, and necroptosis.

7.
Elife ; 82019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31793433

RESUMEN

During human immunodeficiency virus-1 (HIV-1) infection lymphoid organ follicular dendritic cells (FDCs) serve as a reservoir for infectious virus and an obstacle to curative therapies. Here, we identify a subset of lymphoid organ sinus lining macrophage (SMs) that provide a cell-cell contact portal, which facilitates the uptake of HIV-1 viral-like particles (VLPs) by FDCs and B cells in mouse lymph node. Central for portal function is the bridging glycoprotein MFG-E8. Using a phosphatidylserine binding domain and an RGD motif, MFG-E8 helps target HIV-1 VLPs to αv integrin bearing SMs. Lack of MFG-E8 or integrin blockade severely limits HIV-1 VLP spread onto FDC networks. Direct SM-FDC virion transfer also depends upon short-lived FDC network abutment, likely triggered by SCSM antigen uptake. This provides a mechanism for rapid FDC loading broadening the opportunity for rare, antigen reactive follicular B cells to acquire antigen, and a means for HIV virions to accumulate on the FDC network.


Asunto(s)
Antígenos de Superficie/genética , Células Dendríticas Foliculares/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Proteínas de la Leche/genética , Animales , Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Línea Celular , Células Dendríticas Foliculares/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Integrina alfaV/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Macrófagos/inmunología , Macrófagos/virología , Ratones , Proteínas de la Leche/inmunología
8.
J Exp Med ; 216(8): 1749-1761, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31201207

RESUMEN

Preselection thymocytes are normally retained in the thymic cortex, but the mechanisms responsible remain incompletely understood. We now report that deletion of genes encoding the E-protein transcription factors E2A and HEB disorders chemokine receptor expression on developing thymocytes to allow escape of preselection TCR-CD8+ thymocytes into the periphery. We document that CXCR4 expression normally anchors preselection thymocytes to the thymic cortex via interaction with its ligand CXCL12 on cortical thymic epithelial cells, and that disruption of CXCR4-CXCL12 engagements release preselection thymocytes from the thymic cortex. We further document that CXCR4 expression must be extinguished by TCR-mediated positive selection signals to allow migration of TCR-signaled thymocytes out of the thymic cortex into the medulla. Thus, E-protein transcription factors regulate the ordered expression pattern of chemokine receptors on developing thymocytes, and the interaction of the chemokine receptor CXCR4 with its ligand adheres TCR-unsignaled preselection thymocytes to the thymic cortex.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Receptores CXCR4/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Antígenos CD8/metabolismo , Diferenciación Celular/genética , Quimiocina CXCL12/metabolismo , Células Epiteliales/metabolismo , Humanos , Linfopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR4/genética , Transducción de Señal/genética
9.
Mol Cell ; 73(3): 391-392, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735651

RESUMEN

IRGM is a risk factor for several inflammatory diseases, yet no direct link to immune regulation had been shown. In this issue of Molecular Cell, Mehto et al. (2019) report that IRGM limits NLRP3 inflammasome activation-by both direct inhibition of NLRP3/ASC oligomerization and selective autophagic destruction of NLRP3/ASC.


Asunto(s)
Autofagia , Enfermedad de Crohn , Proteínas de Unión al GTP , Humanos , Inmunidad Innata , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Factores de Riesgo
10.
J Immunol ; 202(5): 1510-1520, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30683698

RESUMEN

Macrophages exist as innate immune subsets that exhibit phenotypic heterogeneity and functional plasticity. Their phenotypes are dictated by inputs from the tissue microenvironment. G-protein-coupled receptors are essential in transducing signals from the microenvironment, and heterotrimeric Gα signaling links these receptors to downstream effectors. Several Gαi-coupled G-protein-coupled receptors have been implicated in macrophage polarization. In this study, we use genetically modified mice to investigate the role of Gαi2 on inflammasome activity and macrophage polarization. We report that Gαi2 in murine bone marrow-derived macrophages (BMDMs) regulates IL-1ß release after activation of the NLRP3, AIM2, and NLRC4 inflammasomes. We show this regulation stems from the biased polarity of Gαi2 deficient (Gnai2 -/-) and RGS-insensitive Gαi2 (Gnai2 G184S/G184S) BMDMs. We determined that although Gnai2 G184S/G184S BMDMs (excess Gαi2 signaling) have a tendency toward classically activated proinflammatory (M1) phenotype, Gnai2-/- BMDMs (Gαi2 deficient) are biased toward alternatively activated anti-inflammatory (M2) phenotype. Finally, we find that Gαi2-deficient macrophages have increased Akt activation and IFN-ß production but defects in ERK1/2 and STAT3 activation after LPS stimulation. Gαi2-deficient macrophages also exhibit increased STAT6 activation after IL-4 stimulation. In summary, our data indicates that excess Gαi2 signaling promotes an M1 macrophage phenotype, whereas Gαi2 signaling deficiency promotes an M2 phenotype. Understanding Gαi2-mediated effects on macrophage polarization may bring to light insights regarding disease pathogenesis and the reprogramming of macrophages for the development of novel therapeutics.


Asunto(s)
Citocinas/biosíntesis , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Animales , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
11.
Cell Death Dis ; 9(9): 904, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185776

RESUMEN

The molecular mechanisms underlying the severe lung pathology that occurs during SARS-CoV infections remain incompletely understood. The largest of the SARS-CoV accessory protein open reading frames (SARS 3a) oligomerizes, dynamically inserting into late endosomal, lysosomal, and trans-Golgi-network membranes. While previously implicated in a non-inflammatory apoptotic cell death pathway, here we extend the range of SARS 3a pathophysiologic targets by examining its effects on necrotic cell death pathways. We show that SARS 3a interacts with Receptor Interacting Protein 3 (Rip3), which augments the oligomerization of SARS 3a helping drive necrotic cell death. In addition, by inserting into lysosomal membranes SARS 3a triggers lysosomal damage and dysfunction. Consequently, Transcription Factor EB (TFEB) translocates to the nucleus increasing the transcription of autophagy- and lysosome-related genes. Finally, SARS 3a activates caspase-1 either directly or via an enhanced potassium efflux, which triggers NLRP3 inflammasome assembly. In summary, Rip3-mediated oligomerization of SARS 3a causes necrotic cell death, lysosomal damage, and caspase-1 activation-all likely contributing to the clinical manifestations of SARS-CoV infection.


Asunto(s)
Necrosis/virología , Sistemas de Lectura Abierta/genética , Síndrome Respiratorio Agudo Grave/patología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Células A549 , Apoptosis/fisiología , Autofagia/fisiología , Línea Celular , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Inflamasomas/metabolismo , Membranas Intracelulares/patología , Membranas Intracelulares/virología , Lisosomas/metabolismo , Lisosomas/patología , Lisosomas/virología , Necrosis/metabolismo , Necrosis/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Síndrome Respiratorio Agudo Grave/virología
12.
Sci Rep ; 7(1): 4156, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28646160

RESUMEN

Thymocyte and T cell trafficking relies on signals initiated by G-protein coupled receptors. To address the importance of the G-proteins Gαi2 and Gαi3 in thymocyte and T cell function, we developed several mouse models. Gαi2 deficiency in hematopoietic progenitors led to a small thymus, a double negative (DN)1/DN2 thymocyte transition block, and an accumulation of mature single positive (SP) thymocytes. Loss at the double positive (DP) stage of thymocyte development caused an increase in mature cells within the thymus. In both models an abnormal distribution of memory and naïve CD4 T cells occurred, and peripheral CD4 and CD8 T cells had reduced chemoattractant responses. The loss of Gαi3 had no discernable impact, however the lack of both G-proteins commencing at the DP stage caused a severe T cell phenotype. These mice lacked a thymic medullary region, exhibited thymocyte retention, had a peripheral T cell deficiency, and lacked T cell chemoattractant responses. Yet a noteworthy population of CD4+PD-1+CXCR5+/- cells resided in the spleen of these mice likely due to a loss of regulatory T cell function. Our results delineate a role for Gαi2 in early thymocyte development and for Gαi2/3 in multiple aspects of T cell biology.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Movimiento Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Receptor de Muerte Celular Programada 1/metabolismo , Receptores CXCR5/metabolismo , Bazo/citología , Timocitos/citología , Animales , Compartimento Celular , Proliferación Celular , Quimiocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Timocitos/metabolismo
13.
Sci Rep ; 7: 41258, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28117437

RESUMEN

Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by autoantibody production. Mesenchymal stem cells (MSCs) ameliorate SLE symptoms by targeting T cells, whereas the mechanisms of their efficacy remain incompletely understood. In this study, we show that transfer of human MSCs increased MRL.Faslpr mouse survival, decreased T cell infiltration in the kidneys, and reduced T cell cytokine expression. In vitro, allogeneic mouse MSCs inhibited MRL.Faslpr T cell proliferation and cytokine production. Time-lapse imaging revealed that MSCs recruited MRL.Faslpr T cells establishing long-lasting cellular contacts by enhancing T cell VCAM-1 expression in a CCL2-dependent manner. In contrast, CCL2 deficient MSCs did not induce T cell migration and VCAM-1 expression, resulting in insufficient cell-cell contact. Consequently, CCL2 deficient MSCs did not inhibit IFN-γ production by T cells and upon transfer no longer prolonged survival of MRL.Faslpr mice. Taken together, our imaging study demonstrates that CCL2 enables the prolonged MSC-T cell interactions needed for sufficient suppression of autoreactive T cells and helps to understand how MSCs ameliorate symptoms in lupus-prone MRL.Faslpr mice.


Asunto(s)
Comunicación Celular , Quimiocina CCL2/deficiencia , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Células Madre Mesenquimatosas/metabolismo , Linfocitos T/metabolismo , Animales , Movimiento Celular , Humanos , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Solubilidad , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Mol Cell Oncol ; 3(5): e1078923, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27857968

RESUMEN

Paradoxically, both anticancer immunosurveillance and tumor progression have been associated with intact autophagy, which is regulated by the target of rapamycin (Tor1). Here, we describe the potential impact on the design of cancer therapeutics of a newly described highly conserved post-transcriptional mechanism whereby Tor regulates autophagy.

15.
J Immunol ; 196(2): 846-56, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26667172

RESUMEN

Many intracellular pathogens cause disease by subverting macrophage innate immune defense mechanisms. Intracellular pathogens actively avoid delivery to or directly target lysosomes, the major intracellular degradative organelle. In this article, we demonstrate that activator of G-protein signaling 3 (AGS3), an LPS-inducible protein in macrophages, affects both lysosomal biogenesis and activity. AGS3 binds the Gi family of G proteins via its G-protein regulatory (GoLoco) motif, stabilizing the Gα subunit in its GDP-bound conformation. Elevated AGS3 levels in macrophages limited the activity of the mammalian target of rapamycin pathway, a sensor of cellular nutritional status. This triggered the nuclear translocation of transcription factor EB, a known activator of lysosomal gene transcription. In contrast, AGS3-deficient macrophages had increased mammalian target of rapamycin activity, reduced transcription factor EB activity, and a lower lysosomal mass. High levels of AGS3 in macrophages enhanced their resistance to infection by Burkholderia cenocepacia J2315, Mycobacterium tuberculosis, and methicillin-resistant Staphylococcus aureus, whereas AGS3-deficient macrophages were more susceptible. We conclude that LPS priming increases AGS3 levels, which enhances lysosomal function and increases the capacity of macrophages to eliminate intracellular pathogens.


Asunto(s)
Infecciones Bacterianas/inmunología , Proteínas Portadoras/inmunología , Lisosomas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Citometría de Flujo , Inhibidores de Disociación de Guanina Nucleótido , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño
16.
J Immunol ; 195(5): 2090-102, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26232433

RESUMEN

Resistance to inhibitors of cholinesterase 8A (Ric-8A) is a highly evolutionarily conserved cytosolic protein initially identified in Caenorhabditis elegans, where it was assigned a regulatory role in asymmetric cell divisions. It functions as a guanine nucleotide exchange factor for Gαi, Gαq, and Gα12/13 and as a molecular chaperone required for the initial association of nascent Gα subunits with cellular membranes in embryonic stem cell lines. To test its role in hematopoiesis and B lymphocytes specifically, we generated ric8 (fl/fl) vav1-cre and ric8 (fl/fl) mb1-cre mice. The major hematopoietic cell lineages developed in the ric8 (fl/fl) vav1-cre mice, notwithstanding severe reduction in Gαi2/3, Gαq, and Gα13 proteins. B lymphocyte-specific loss of Ric-8A did not compromise bone marrow B lymphopoiesis, but splenic marginal zone B cell development failed, and B cells underpopulated lymphoid organs. The ric8 (fl/fl) mb1-cre B cells exhibited poor responses to chemokines, abnormal trafficking, improper in situ positioning, and loss of polarity components during B cell differentiation. The ric8 (fl/fl) mb1-cre mice had a severely disrupted lymphoid architecture and poor primary and secondary Ab responses. In B lymphocytes, Ric-8A is essential for normal Gα protein levels and is required for B cell differentiation, trafficking, and Ab responses.


Asunto(s)
Linfocitos B/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/inmunología , Factores de Intercambio de Guanina Nucleótido/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Animales , Linfocitos B/metabolismo , Western Blotting , Calcio/inmunología , Calcio/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Inmunidad Humoral/genética , Inmunidad Humoral/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/inmunología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Imagen de Lapso de Tiempo
17.
Elife ; 42015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26258881

RESUMEN

The HIV-1 envelope protein gp120 is both the target of neutralizing antibodies and a major focus of vaccine efforts; however how it is delivered to B cells to elicit an antibody response is unknown. Here, we show that following local gp120 injection lymph node (LN) SIGN-R1(+) sinus macrophages located in interfollicular pockets and underlying SIGN-R1(+) macrophages form a cellular network that rapidly captures gp120 from the afferent lymph. In contrast, two other antigens, phycoerythrin and hen egg lysozyme, were not captured by these cells. Intravital imaging of mouse LNs revealed persistent, but transient interactions between gp120 bearing interfollicular network cells and both trafficking and LN follicle resident gp120 specific B cells. The gp120 specific, but not the control B cells repetitively extracted gp120 from the network cells. Our findings reveal a specialized LN antigen delivery system poised to deliver gp120 and likely other pathogen derived glycoproteins to B cells.


Asunto(s)
Presentación de Antígeno , Linfocitos B/inmunología , Moléculas de Adhesión Celular/análisis , Proteína gp120 de Envoltorio del VIH/inmunología , Lectinas Tipo C/análisis , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Receptores de Superficie Celular/análisis , Animales , Proteínas del Huevo/administración & dosificación , Proteínas del Huevo/inmunología , Proteína gp120 de Envoltorio del VIH/administración & dosificación , Inmunofenotipificación , Macrófagos/química , Ratones Endogámicos C57BL , Muramidasa/administración & dosificación , Muramidasa/inmunología , Ficoeritrina/administración & dosificación , Ficoeritrina/inmunología
18.
Nat Cell Biol ; 17(7): 930-942, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26098573

RESUMEN

Autophagy is an essential eukaryotic pathway requiring tight regulation to maintain homeostasis and preclude disease. Using yeast and mammalian cells, we report a conserved mechanism of autophagy regulation by RNA helicase RCK family members in association with the decapping enzyme Dcp2. Under nutrient-replete conditions, Dcp2 undergoes TOR-dependent phosphorylation and associates with RCK members to form a complex with autophagy-related (ATG) mRNA transcripts, leading to decapping, degradation and autophagy suppression. Simultaneous with the induction of ATG mRNA synthesis, starvation reverses the process, facilitating ATG mRNA accumulation and autophagy induction. This conserved post-transcriptional mechanism modulates fungal virulence and the mammalian inflammasome, the latter providing mechanistic insight into autoimmunity reported in a patient with a PIK3CD/p110δ gain-of-function mutation. We propose a dynamic model wherein RCK family members, in conjunction with Dcp2, function in controlling ATG mRNA stability to govern autophagy, which in turn modulates vital cellular processes affecting inflammation and microbial pathogenesis.


Asunto(s)
Autofagia/genética , ARN Helicasas DEAD-box/genética , Estabilidad del ARN/genética , Proteínas de Saccharomyces cerevisiae/genética , Animales , Autoinmunidad/genética , Línea Celular Tumoral , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Cryptococcus neoformans/genética , Cryptococcus neoformans/metabolismo , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Femenino , Regulación Fúngica de la Expresión Génica , Células HeLa , Humanos , Immunoblotting , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
19.
J Immunol ; 194(5): 2128-39, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25617475

RESUMEN

Chemokines engage B lymphocyte surface receptors, triggering heterotrimeric G protein Gαi subunit guanine nucleotide exchange. RGS proteins limit the duration that Gαi subunits remain GTP bound, and the loss of an individual RGS protein typically enhances chemokine receptor signaling. In this study, we show that B cells carrying a Gαi2 (G184S/G184S) mutation that disables all RGS protein/Gαi2 interactions exhibit an unexpectedly severe reduction in chemokine receptor signaling. The Gαi2 (G184S/G184S) B cells have markedly elevated basal calcium levels, but poor chemokine-induced increases, enhanced nonspecific migration, but extremely poor chemotaxis. In striking contrast, the Gαi2 (G184S/G184S) B cells exhibited enhanced sensitivity to sphingosine 1-phosphate (S1P). S1P elicited heightened intracellular calcium responses and enhanced S1P-triggered cell migration. Mice with the Gαi2 (G184S/G184S) mutation displayed excessive numbers of germinal center-like structures; abnormal serum Ig profiles; and aberrant B lymphocyte trafficking. These findings establish an essential role for RGS proteins in B cell chemoattractant signaling and for the proper position of B lymphocytes in lymphoid organs.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Proteínas RGS/metabolismo , Bazo/metabolismo , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/inmunología , Sitios de Unión , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Calcio/inmunología , Calcio/metabolismo , Quimiocinas/farmacología , Femenino , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Regulación de la Expresión Génica , Centro Germinal/citología , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Centro Germinal/metabolismo , Lisofosfolípidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Cultivo Primario de Células , Unión Proteica , Proteínas RGS/genética , Proteínas RGS/inmunología , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/farmacología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
20.
Immunol Cell Biol ; 93(1): 11-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25385065

RESUMEN

Autophagy is a major cellular pathway, which at basal levels regulates and maintains the cytoplasmic environment through the capture, isolation and digestion of intracellular materials in a specialized structure called an autophagosome. The unique ability of autophagy to degrade large targets, such as damaged and surplus organelles, intracellular microbes and protein aggregates, has made it a prime focus in inflammation and microbial research. Indeed, autophagy has been shown to be involved in a number of infectious and inflammatory pathologies, by which it may confer protection against intracellular microbes, be targeted by microbes for evasion or be hijacked for microbe biogenesis. In addition, autophagy helps regulate the intracellular and global immune response to both extracellular and intracellular pathogens. Here we review the current literature on the interactions between autophagy and HIV among different immune cells and discuss new research that re-emphasizes the role of inflammation in HIV-mediated CD4(+) T cell death.


Asunto(s)
Autofagia/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Macrófagos/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia , Beclina-1 , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Células Dendríticas/patología , Células Dendríticas/virología , Regulación de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/patología , Infecciones por VIH/virología , Humanos , Evasión Inmune , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/virología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Macrófagos/patología , Macrófagos/virología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Fagosomas/genética , Fagosomas/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Transducción de Señal , Ubiquitina/genética , Ubiquitina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA