Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 7(71): eabh1873, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35622904

RESUMEN

T cells become functionally exhausted in tumors, limiting T cell-based immunotherapies. Although several transcription factors regulating the exhausted T (Tex) cell differentiation are known, comparatively little is known about the regulators of Tex cell survival. Here, we reported that the regulator of G protein signaling 16 (Rgs-16) suppressed Tex cell survival in tumors. By performing lineage tracing using reporter mice in which mCherry marked Rgs16-expressing cells, we identified that Rgs16+CD8+ tumor-infiltrating lymphocytes (TILs) were terminally differentiated, expressed low levels of T cell factor 1 (Tcf1), and underwent apoptosis as early as 6 days after the onset of Rgs16 expression. Rgs16 deficiency inhibited CD8+ T cell apoptosis and promoted antitumor effector functions of CD8+ T cells. Furthermore, Rgs16 deficiency synergized with programmed cell death protein 1 (PD-1) blockade to enhance antitumor CD8+ T cell responses. Proteomics revealed that Rgs16 interacted with the scaffold protein IQGAP1, suppressed the recruitment of Ras and B-Raf, and inhibited Erk1 activation. Rgs16 deficiency enhanced antitumor CD8+ TIL survival in an Erk1-dependent manner. Loss of function of Erk1 decreased antitumor functions of Rgs16-deficient CD8+ T cells. RGS16 mRNA expression levels in CD8+ TILs of patients with melanoma negatively correlated with genes associated with T cell stemness, such as SELL, TCF7, and IL7R, and predicted low responses to PD-1 blockade. This study uncovers Rgs16 as an inhibitor of Tex cell survival in tumors and has implications for improving T cell-based immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Proteínas RGS/inmunología , Animales , Diferenciación Celular , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor , Ratones
2.
Front Immunol ; 13: 1065927, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591293

RESUMEN

Background: Alterations in lipid metabolism promote tumor progression. However, the role of lipid metabolism in the occurrence and development of gastric cancer have not been fully clarified. Method: Here, genes that are related to fatty acid metabolism and differentially-expressed between normal and gastric cancer tissues were identified in the TCGA-STAD cohort. The intersection of identified differentially-expressed genes with Geneset was determined to obtain 78 fatty acid metabolism-related genes. The ConsensusClusterPlus R package was used to perform differentially-expressed genes, which yielded divided two gastric cancer subtypes termed cluster 1 and cluster 2. Results: Patients in cluster 2 was found to display poorer prognosis than patients in cluster 1. Using machine learning method to select 8 differentially expressed genes among subtypes to construct fatty acid prognostic risk score model (FARS), which was found to display good prognostic efficacy. We also identified that certain anticancer drugs, such as bortezomib, elesclomol, GW843682X, and nilotinib, showed significant sensitivity in the high FARS score group. RGS2 was selected as the core gene upon an analysis of the gastric cancer single-cell, and Western blotting and immunofluorescence staining results revealed high level of expression of this gene in gastric cancer cells. The results of immunohistochemical staining showed that a large amount of RGS2 was deposited in the stroma in gastric cancer. A pan-cancer analysis also revealed a significant association of RGS2 with TMB, TIDE, and CD8+ T-cell infiltration in other cancer types as well. RGS2 may thus be studied further as a new target for immunotherapy in future studies on gastric cancer. Conclusion: In summary, the FARS model developed here enhances our understanding of lipid metabolism in the TME in gastric cancer, and provides a theoretical basis for predicting tumor prognosis and clinical treatment.


Asunto(s)
Biomarcadores de Tumor , Ácidos Grasos , Proteínas RGS , Neoplasias Gástricas , Microambiente Tumoral , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Western Blotting , Ácidos Grasos/genética , Ácidos Grasos/inmunología , Ácidos Grasos/metabolismo , Proteínas RGS/genética , Proteínas RGS/inmunología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Pronóstico , Aprendizaje Automático
3.
Neurosci Lett ; 768: 136374, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34852285

RESUMEN

Macrophage-dominated inflammation by the activation of Toll-like receptor (TLR) pathway leads to neurological disruption after spinal cord injury (SCI). Regulator of G-protein signaling 1 (RGS1) is reported to be a regulator in inflammation. The present study thus purposes to identify the unknown role of RGS1 mediating TLR on inflammation post SCI. A mouse model of traumatic SCI was established by a mechanical trauma at T10. The mice underwent SCI and a macrophage line activated by lipopolysaccharide (LPS) were treated with shRNA-RGS1 to elucidate the role of RGS1 in inflammatory progression. The inflammatory factors were measured, and the degree of histology and function protection were determined. The expression levels of RGS1, myeloid differentiation primary response protein 88 (Myd88), (TIR-domain-containing adaptor inducing interferon-ß (TRIF), p38, metalloproteinase (MMP)-2, and MMP-9 were determined. RGS1 was robustly increased both in LPS-activated macrophage and SCI mice. The TLR signaling pathway-induced inflammation was suppressed by RGS1 knockdown. shRNA-mediated silence of RGS1 was exhibited a prominent decrease in TNF-α, IL-1ß and IL-6 via TLR/TRIF/ nuclear factor kappa-B (NF-κB) axis. Depletion of RGS1 also inhibited MMP-induced tissue degradation via MAPK-p38 pathway in SCI mice. Moreover, suppression of RGS1 improved spinal cord histology and function recovery. These findings suggest that RGS1 regulates inflammation and tissue disruption via TLR/TRIF/NF-κB signaling pathway in mice with SCI, thereby explaining a novel target that regulates macrophage inflammation post SCI.


Asunto(s)
Inflamación/inmunología , Macrófagos/inmunología , Proteínas RGS/inmunología , Transducción de Señal/inmunología , Traumatismos de la Médula Espinal/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , FN-kappa B/metabolismo , Células RAW 264.7 , Proteínas RGS/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
4.
Front Immunol ; 12: 772288, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34912341

RESUMEN

Seasonal influenza epidemics represent a significant global health threat. The exacerbated immune response triggered by respiratory influenza virus infection causes severe pulmonary damage and contributes to substantial morbidity and mortality. Regulator of G-protein signaling 10 (RGS10) belongs to the RGS protein family that act as GTPase activating proteins for heterotrimeric G proteins to terminate signaling pathways downstream of G protein-coupled receptors. While RGS10 is highly expressed in immune cells, in particular monocytes and macrophages, where it has strong anti-inflammatory effects, its physiological role in the respiratory immune system has not been explored yet. Here, we show that Rgs10 negatively modulates lung immune and inflammatory responses associated with severe influenza H1N1 virus respiratory infection in a mouse model. In response to influenza A virus challenge, mice lacking RGS10 experience enhanced weight loss and lung viral titers, higher mortality and significantly faster disease onset. Deficiency of Rgs10 upregulates the levels of several proinflammatory cytokines and chemokines and increases myeloid leukocyte accumulation in the infected lung, markedly neutrophils, monocytes, and inflammatory monocytes, which is associated with more pronounced lung damage. Consistent with this, influenza-infected Rgs10-deficent lungs contain more neutrophil extracellular traps and exhibit higher neutrophil elastase activities than wild-type lungs. Overall, these findings propose a novel, in vivo role for RGS10 in the respiratory immune system controlling myeloid leukocyte infiltration, viral clearance and associated clinical symptoms following lethal influenza challenge. RGS10 also holds promise as a new, potential therapeutic target for respiratory infections.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/inmunología , Neumonía/inmunología , Proteínas RGS/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/inmunología , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Neumonía/patología , Neumonía/virología , Proteínas RGS/genética
5.
Front Immunol ; 12: 718359, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867947

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with widespread inflammation, immune dysregulation, and is associated with the generation of destructive anti-DNA autoantibodies. We have shown previously the immune modulatory properties of pCons peptide in the induction of both CD4+ and CD8+ regulatory T cells which can in turn suppress development of the autoimmune disease in (NZB/NZW) F1 (BWF1) mice, an established model of lupus. In the present study, we add novel protein information and further demonstrate the molecular and cellular phenotypes of pCons-induced CD4+ and CD8+ Treg subsets. Flow cytometry analyses revealed that pCons induced CD8+ Treg cells with the following cell surface molecules: CD25highCD28high and low subsets (shown earlier), CD62Lhigh, CD122low, PD1low, CTLA4low, CCR7low and 41BBhigh. Quantitative real-time PCR (qRT-PCR) gene expression analyses revealed that pCons-induced CD8+ Treg cells downregulated the following several genes: Regulator of G protein signaling (RGS2), RGS16, RGS17, BAX, GPT2, PDE3b, GADD45ß and programmed cell death 1 (PD1). Further, we confirmed the down regulation of these genes by Western blot analyses at the protein level. To our translational significance, we showed herein that pCons significantly increased the percentage of CD8+FoxP3+ T cells and further increased the mean fluorescence intensity (MFI) of FoxP3 when healthy peripheral blood mononuclear cells (PBMCs) are treated with pCons (10 µg/ml, for 24-48 hours). In addition, we found that pCons reduced apoptosis in CD4+ and CD8+ T cells and B220+ B cells of BWF1 lupus mice. These data suggest that pCons stimulates cellular, immunological, and molecular changes in regulatory T cells which in turn protect against SLE autoimmunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Péptidos/genética , Péptidos/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/metabolismo , Apoptosis/genética , Apoptosis/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Expresión Génica , Voluntarios Sanos , Humanos , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos NZB , Persona de Mediana Edad , Péptidos/administración & dosificación , Proteínas RGS/genética , Proteínas RGS/inmunología , Adulto Joven
6.
Mol Immunol ; 134: 236-246, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33836352

RESUMEN

Rheumatoid arthritis (RA) is the most common inflammatory arthropathy, with evidence pointing to an immune-mediated etiology that propagates chronic inflammation. Although targeted immune therapeutics and aggressive treatment strategies have substantially improved, a complete understanding of the associated pathological mechanisms of the disease remains elusive. This study aimed at investigating whether regulator of G protein signaling 10 (RGS10) could affect rheumatoid arthritis (RA) pathology by regulating the immune response. A DBA/J1 mouse model of RA was established and evaluated for disease severity. RGS10 expression was inhibited by adeno-associated virus in vivo. Moreover, small interfering RNA was used to downregulate RGS10 expression in raw 264.7 cells in vitro. Results showed that RGS10 inhibition augmented RA severity, and attenuated the increase in expression of inflammatory factors. Furthermore, activated NF-κB signaling pathways were detected following RGS10 inhibition. These results revealed that RGS10 inhibition directly aggravated the RA pathological process by activating the NF-κB signaling pathway. Therefore, RGS10 is a promising novel therapeutic target for RA treatment with a potential clinical impact.


Asunto(s)
Artritis Reumatoide/inmunología , FN-kappa B/metabolismo , Proteínas RGS/inmunología , Transducción de Señal/inmunología , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Progresión de la Enfermedad , Ratones , Ratones Endogámicos DBA , Células RAW 264.7 , Proteínas RGS/metabolismo
7.
Anal Chem ; 91(5): 3327-3335, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30701963

RESUMEN

Lung cancer is the primary cause of cancer-associated mortality worldwide, which makes the identification of reliable lung cancer biomarkers a pressing need for early diagnosis and prognosis. RGS11, which is a regulator of G-protein signaling and also a lung cancer biomarker, plays an important role in cancer-related metastasis. However, trace levels of RGS11 (in the range of pg/mL) in serum samples make it difficult to quantify using currently available enzyme-linked immunosorbent assay (ELISA) kits and, therefore, this hinders progress in the discovery of new approaches for treating lung cancer. The aim of this study is to develop a rapid, sensitive, and reliable platform for the detection of RGS11 lung cancer biomarker based on a suspension immunoassay coupled with an isothermal exponential amplification strategy. Our study was initiated by the functionalization of magnetic beads with anti-RGS11 antibodies (Ab-MB) by EDC (1-ethyl-3-(3-(dimethylamino)propyl)-carbodiimide)/NHS ( N-hydroxysulfosuccinimide) activation. Ab-MB served as a sensing probe for the competitive immunorecognitions between known concentrations of His-tag RGS11 and unknown concentrations of target RGS11 in serum. The reporter anti-His antibodies, which were modified with primers that induced an isothermal exponential amplification reaction, were subsequently introduced to the reaction mixture that resulted in the formation of immunosandwich complexes. The exponentially amplified DNA duplex that was intercalated with SYBR Green was designated as a signal reporter for the assessment of RGS11 in an inversely proportional relationship. The sensing platform was excellent for the determination of RGS11 with an exceptional detection limit of 148 fg/mL and a linear dynamic range of 0.1-10 pg/mL using a minimal sample volume (20 µL) and with a reaction time of 1.5 h. In addition, we challenged the sensing platform with RGS11-spiked samples (in 2× diluted serum), and an acceptable recovery rate (>90%) was observed. Finally, 24 clinical samples acquired from patients with advanced lung cancer (C), inflammation (I), and heart failure (H) were analyzed by this newly developed sensing platform and a commercial ELISA kit for validation. This sensing platform has potential in biomedical applications for clinically diagnosing liquid biopsy samples for patients with lung cancer. Moreover, the universal design of our proposed system is easily adapted to detect any other protein if a His-tag recombinant protein is available.


Asunto(s)
Biomarcadores de Tumor/sangre , Inmunoensayo/métodos , Neoplasias Pulmonares/diagnóstico , Proteínas RGS/sangre , Anticuerpos Inmovilizados/química , Anticuerpos Inmovilizados/inmunología , Reacciones Antígeno-Anticuerpo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Histidina/genética , Histidina/inmunología , Histidina/metabolismo , Humanos , Límite de Detección , Neoplasias Pulmonares/metabolismo , Magnetismo , Técnicas de Amplificación de Ácido Nucleico , Oligopéptidos/genética , Oligopéptidos/inmunología , Oligopéptidos/metabolismo , Proteínas RGS/genética , Proteínas RGS/inmunología
8.
Biochem Biophys Res Commun ; 516(3): 1039-1045, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-28698136

RESUMEN

Heterotrimeric G proteins composed of Gα, Gß and Gγ subunits are evolutionarily conserved signaling modules involved in diverse biological processes in plants and animals. The role and action of Gα remain largely enigmatic in plant innate immunity. We have recently demonstrated that Arabidopsis Gα (GPA1) is a key component of a new immune signaling pathway activated by bacteria-secreted proteases. Here we show that GPA1 is also involved in the signaling network of Arabidopsis in response to the bacterial flagellin epitope flg22. Specifically, GPA1 plays a pivotal role in an immune pathway involving the flg22 receptor FLS2, co-receptor BAK1, Regulator of G Signaling 1 (RGS1), and Arabidopsis Gß (AGB1), in which flg22 elicits GPA1/AGB1 dissociation from the FLS2/BAK1/RGS1 receptor complex. Consequently, we observed flg22-induced degradation of FLS2, BAK1 and RGS1 but not GPA1 or AGB1. We also found that GPA1 constitutively interacts with the NADPH oxidase RbohD to potentiate flg22-induced ROS burst independently of the central cytoplasmic kinase BIK1. Taken together, our work sheds multiple novel insights into the functions and regulatory mechanisms of GPA1 in Arabidopsis innate immunity.


Asunto(s)
Proteínas de Arabidopsis/inmunología , Arabidopsis/inmunología , Flagelina/inmunología , Subunidades alfa de la Proteína de Unión al GTP/inmunología , Inmunidad Innata/inmunología , Transducción de Señal/inmunología , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Epítopos/inmunología , Flagelina/química , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/inmunología , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Inmunidad Innata/genética , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , NADPH Oxidasas/metabolismo , Plantas Modificadas Genéticamente , Unión Proteica , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas RGS/genética , Proteínas RGS/inmunología , Proteínas RGS/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética
9.
J Biol Chem ; 293(13): 4752-4766, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382719

RESUMEN

Heterotrimeric G protein complexes are molecular switches relaying extracellular signals sensed by G protein-coupled receptors (GPCRs) to downstream targets in the cytoplasm, which effect cellular responses. In the plant heterotrimeric GTPase cycle, GTP hydrolysis, rather than nucleotide exchange, is the rate-limiting reaction and is accelerated by a receptor-like regulator of G signaling (RGS) protein. We hypothesized that posttranslational modification of the Gα subunit in the G protein complex regulates the RGS-dependent GTPase cycle. Our structural analyses identified an invariant phosphorylated tyrosine residue (Tyr166 in the Arabidopsis Gα subunit AtGPA1) located in the intramolecular domain interface where nucleotide binding and hydrolysis occur. We also identified a receptor-like kinase that phosphorylates AtGPA1 in a Tyr166-dependent manner. Discrete molecular dynamics simulations predicted that phosphorylated Tyr166 forms a salt bridge in this interface and potentially affects the RGS protein-accelerated GTPase cycle. Using a Tyr166 phosphomimetic substitution, we found that the cognate RGS protein binds more tightly to the GDP-bound Gα substrate, consequently reducing its ability to accelerate GTPase activity. In conclusion, we propose that phosphorylation of Tyr166 in AtGPA1 changes the binding pattern with AtRGS1 and thereby attenuates the steady-state rate of the GTPase cycle. We coin this newly identified mechanism "substrate phosphoswitching."


Asunto(s)
Proteínas de Arabidopsis/inmunología , Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas RGS/inmunología , Sustitución de Aminoácidos , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Mutación Missense , Fosforilación , Proteínas RGS/genética , Tirosina/genética , Tirosina/metabolismo
10.
Am J Respir Cell Mol Biol ; 58(1): 89-98, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28853915

RESUMEN

A cardinal feature of asthma is airway hyperresponsiveness (AHR) to spasmogens, many of which activate G protein-coupled receptors (GPCRs) on airway smooth muscle (ASM) cells. Asthma subtypes associated with allergy are characterized by eosinophilic inflammation in the lung due to the type 2 immune response to allergens and proinflammatory mediators that promote AHR. The degree to which intrinsic abnormalities of ASM contribute to this phenotype remains unknown. The regulators of G protein signaling (RGS) proteins are a large group of intracellular proteins that inhibit GPCR signaling pathways. RGS2- and RGS5-deficient mice develop AHR spontaneously. Although RGS4 is upregulated in ASM from patients with severe asthma, the effects of increased RGS4 expression on AHR in vivo are unknown. Here, we examined the impact of forced RGS4 overexpression in lung on AHR using transgenic (Tg) mice. Tg RGS4 was expressed in bronchial epithelium and ASM in vivo, and protein expression in lung was increased at least 4-fold in Tg mice compared with wild-type (WT) mice. Lung slices from Tg mice contracted less in response to the m3 muscarinic receptor agonist methacholine compared with the WT, although airway resistance in live, unchallenged mice of both strains was similar. Tg mice were partially protected against AHR induced by fungal allergen challenge due to weakened contraction signaling in ASM and reduced type 2 cytokine (IL-5 and IL-13) levels in Tg mice compared with the WT. These results provide support for the hypothesis that increasing RGS4 expression and/or function could be a viable therapeutic strategy for asthma.


Asunto(s)
Asma/inmunología , Bronquios/inmunología , Regulación de la Expresión Génica/inmunología , Pulmón/inmunología , Proteínas RGS/inmunología , Mucosa Respiratoria/inmunología , Animales , Asma/genética , Asma/patología , Bronquios/patología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Pulmón/patología , Ratones , Ratones Transgénicos , Proteínas RGS/genética , Mucosa Respiratoria/patología
11.
J Immunol ; 198(7): 2721-2734, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28235863

RESUMEN

Adaptive immunity depends on mature thymocytes leaving the thymus to enter the bloodstream and the trafficking of T cells through lymphoid organs. Both of these require heterotrimeric Gαi protein signaling, whose intensity and duration are controlled by the regulator of G protein signaling (RGS) proteins. In this study, we show that RGS protein/Gαi2 interactions are essential for normal thymocyte egress, T cell trafficking, and homeostasis. Mature thymocytes with a Gαi2 mutation that disables RGS protein binding accumulated in the perivascular channels of thymic corticomedullary venules. Severe reductions in peripheral naive CD4+ T cells and regulatory T cells occurred. The mutant CD4+ T cells adhered poorly to high endothelial venules and exhibited defects in lymph node entrance and egress. The kinetics of chemokine receptor signaling were disturbed, including chemokine- induced integrin activation. Despite the thymic and lymph node egress defects, sphingosine-1-phosphate signaling was not obviously perturbed. This study reveals how RGS proteins modulate Gαi2 signaling to facilitate thymocyte egress and T cell trafficking.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiotaxis de Leucocito/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Proteínas RGS/inmunología , Timocitos/inmunología , Inmunidad Adaptativa/inmunología , Animales , Citometría de Flujo , Homeostasis/inmunología , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Transducción de Señal/inmunología
12.
Biochem Pharmacol ; 114: 40-52, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27071343

RESUMEN

Leukocyte chemoattractant receptors are members of the G-protein coupled receptor (GPCR) family. Signaling downstream of these receptors directs the localization, positioning and homeostatic trafficking of leukocytes; as well as their recruitment to, and their retention at, inflammatory sites. Ligand induced changes in the molecular conformation of chemoattractant receptors results in the engagement of heterotrimeric G-proteins, which promotes α subunits to undergo GTP/GDP exchange. This results in the functional release of ßγ subunits from the heterotrimers, thereby activating downstream effector molecules, which initiate leukocyte polarization, gradient sensing, and directional migration. Pertussis toxin ADP ribosylates Gαi subunits and prevents chemoattractant receptors from triggering Gαi nucleotide exchange. The use of pertussis toxin revealed the essential importance of Gαi subunit nucleotide exchange for chemoattractant receptor signaling. More recent studies have identified a range of regulatory mechanisms that target these receptors and their associated heterotrimeric G-proteins, thereby helping to control the magnitude, kinetics, and duration of signaling. A failure in these regulatory pathways can lead to impaired receptor signaling and immunopathology. The analysis of mice with targeted deletions of Gαi isoforms as well as some of these G-protein regulatory proteins is providing insights into their roles in chemoattractant receptor signaling.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Sistema Inmunológico/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Humanos , Toxina del Pertussis/farmacología , Proteínas RGS/inmunología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/inmunología
13.
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
14.
J Allergy Clin Immunol ; 135(1): 81-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25226851

RESUMEN

BACKGROUND: Childhood asthma is classified into allergic asthma (AA) and nonallergic asthma (NA), yet both are treated identically, with only partial success. OBJECTIVE: We sought to identify novel immune phenotypes for childhood AA and NA. METHODS: The Clinical Asthma Research Association cohort study includes 275 steroid-naive 4- to 15-year-old German children (healthy control subjects [HCs], patients with AA, and patients with NA). In PBMCs both quantitative and functional analysis of regulatory T (Treg) and TH17 cells (flow cytometry/Treg cell suppression) before/after anti-CD3/CD28, lipid A, and peptidoglycan stimulation were performed. Cytokines and gene expression, as assessed by using Luminex or transcriptomics/quantitative real-time RT-PCR, were analyzed by means of regression analysis. Linear discriminant analysis was applied to discriminate between phenotypes. RESULTS: The 3 phenotypes were immunologically well discriminated by means of microarray and protein analysis with linear discriminant analysis. Patients with AA were characterized by increased Treg cells compared with those in HCs but not those in patients with NA. Treg cells from patients with AA, but not patients with NA, significantly suppressed IL-5, IL-13, and IFN-γ secretion. Patients with AA had decreased expression of chloride intracellular channel 4 (CLIC4) and tuberous sclerosis 1 (TSC1), important innate immunity regulators. Patients with NA were characterized by increased proinflammatory IL-1ß levels, neutrophil counts, and IL-17-shifted immunity. In parallel, expressions of anti-inflammatory IL37, proline-serine-threonine phosphatase-interacting protein 2 (PSTPIP2), and the neutrophil-associated genes CD93, triggering receptor expressed on myeloid cells 1 (TREM1), and regulator of G-protein signaling 13 (RGS13) were increased in patients with NA. A shared TH2 immunity was present in both asthma phenotypes. CONCLUSION: Novel immune-regulatory mechanisms in childhood asthma identified increased Treg cells in patients with AA compared with those in HCs but not those in NA and decreased innate immunity genes for patients with AA, the first potentially indicating a counterregulatory mechanism to suppress cytokines yet not sufficient to control allergic inflammation. Very distinctly, patients with NA showed an IL-17-shifted proinflammatory immunity, promoting neutrophil inflammation and less functional Treg cells. Identification of these unique pathways provides a profound basis for future strategies for individualized prediction of asthma development, disease course, and prevention.


Asunto(s)
Asma/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Adolescente , Niño , Preescolar , Canales de Cloruro/inmunología , Citocinas/inmunología , Proteínas del Citoesqueleto/inmunología , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/inmunología , Fenotipo , Proteínas RGS/inmunología , Receptores de Complemento/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología , Receptor Activador Expresado en Células Mieloides 1 , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/inmunología
15.
Scand J Immunol ; 81(1): 23-30, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25366993

RESUMEN

Immune cells express powerful and harmful effectors that require tight regulation. Heterotrimeric G proteins are critical mediators in translating extracellular signals into cell responses, which need a fine-tuned regulation for the control of cell activation. Regulator of G-protein signalling 16 (RGS16) has been identified as a key factor of G protein-mediated activation in lymphocytes, modulating inflammatory and survival responses of various cell types. However, data about the expression of this regulatory protein in monocytes are scarce, and it has remained unclear whether activation and migration of these cells are regulated by RGS16. In this study, the impact of RGS16 on the production of inflammatory cytokines by activated human monocytes was investigated in vitro using the human promonocytic cell line THP-1 as a model. Gain and loss of function experiments showed that RGS16 overexpression reduces the expression of pro-inflammatory cytokines IL-1ß, IL-6, IL-8 and TNFα, while RGS16 knockdown by RNAi upregulates IL-1ß, IL-6 and TNFα but not IL-8. RGS16 knockdown was also shown to enhance Pam3-mediated induction of the anti-inflammatory cytokine IL-10. Our results indicate that RGS16 restricts the activation-induced pro-inflammatory profile in myeloid cells.


Asunto(s)
Inflamación/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Proteínas RGS/inmunología , Animales , Células de la Médula Ósea , Línea Celular , Humanos , Interleucina-10/biosíntesis , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Interleucina-8/biosíntesis , Lipopéptidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas RGS/biosíntesis , Proteínas RGS/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptor Toll-Like 2/agonistas , Factor de Necrosis Tumoral alfa/biosíntesis
16.
Immunobiology ; 219(9): 653-60, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24775271

RESUMEN

Macrophages play critical roles in innate immune defense by sensing microbes using pattern-recognition receptors. Lipopolysaccharide (LPS) stimulates macrophages via TLR, which leads to activation of downstream signaling cascades. In this study, we investigated the roles of a conserved signaling pathway, Notch signaling, in regulating the downstream signaling cascades of the LPS/TLR4 pathways in macrophages. Using a phospho-proteomic approach and a gamma-secretase inhibitor (GSI) to suppress the processing and activation of Notch signaling, we identified regulator of G protein signaling 19 (RGS19) as a target protein whose phosphorylation was affected by GSI treatment. RGS19 is a guanosine triphosphatase (GTPase)-activating protein that functions to negatively regulate G protein-coupled receptors via Gαi/Gαq-linked signaling. Stimulation of RAW264.7 cells with LPS increased the level of the phosphorylated form of RGS19, while LPS stimulation in the presence of GSI decreased its level. GSI treatment did not alter the mRNA level of rgs19. Treatment with GSI or silencing of rgs19 in macrophages impaired the phosphorylation of Akt Thr(308) upon LPS stimulation. Furthermore, targeted deletion of a DNA-binding protein and binding partner of the Notch receptor, RBP-Jκ/CSL, in macrophages resulted in delayed and decreased Akt phosphorylation. Because the PI3K/Akt pathway regulates cell survival in various cell types, the cell cycle and cell death were assayed upon GSI treatment, phosphatidylinositol 3 kinase (PI3K) inhibitor treatment or silencing of rgs19. GSI treatment resulted in decreased cell populations in the G1 and S phases, while it increased the cell population of cell death. Similarly, silencing of rgs19 resulted in a decreased cell population in the G1 phase and an increased cell population in the subG1 phase. Inhibition of Akt phosphorylation by PI3K inhibitor in LPS-stimulated macrophages increased cell population in G1 phase, suggesting a possible cell cycle arrest. Taken together, these results indicate that Notch signaling positively regulates phosphorylation of Akt, possibly via phosphorylation of RGS19, and inhibition of both molecules affects the cell survival and cell cycle of macrophages upon LPS stimulation.


Asunto(s)
Macrófagos/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas RGS/inmunología , Receptores Notch/inmunología , Transducción de Señal/inmunología , Animales , Apoptosis , Western Blotting , Línea Celular , Electroforesis en Gel Bidimensional , Citometría de Flujo , Lipopolisacáridos/inmunología , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Fosforilación , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
J Allergy Clin Immunol ; 134(2): 451-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24666695

RESUMEN

BACKGROUND: Although eosinophilic inflammation typifies allergic asthma, it is not a prerequisite for airway hyperresponsiveness (AHR), suggesting that underlying abnormalities in structural cells, such as airway smooth muscle (ASM), contribute to the asthmatic diathesis. Dysregulation of procontractile G protein-coupled receptor (GPCR) signaling in ASM could mediate enhanced contractility. OBJECTIVE: We explored the role of a regulator of procontractile GPCR signaling, regulator of G protein signaling 5 (RGS5), in unprovoked and allergen-induced AHR. METHODS: We evaluated GPCR-evoked Ca(2+) signaling, precision-cut lung slice (PCLS) contraction, and lung inflammation in naive and Aspergillus fumigatus-challenged wild-type and Rgs5(-/-) mice. We analyzed lung resistance and dynamic compliance in live anesthetized mice using invasive plethysmography. RESULTS: Loss of RGS5 promoted constitutive AHR because of enhanced GPCR-induced Ca(2+) mobilization in ASM. PCLSs from naive Rgs5(-/-) mice contracted maximally at baseline independently of allergen challenge. RGS5 deficiency had little effect on the parameters of allergic inflammation, including cell counts in bronchoalveolar lavage fluid, mucin production, ASM mass, and subepithelial collagen deposition. Unexpectedly, induced IL-13 and IL-33 levels were much lower in challenged lungs from Rgs5(-/-) mice relative to those seen in wild-type mice. CONCLUSION: Loss of RGS5 confers spontaneous AHR in mice in the absence of allergic inflammation. Because it is selectively expressed in ASM within the lung and does not promote inflammation, RGS5 might be a therapeutic target for asthma.


Asunto(s)
Alérgenos/inmunología , Calcio/inmunología , Pulmón/patología , Miocitos del Músculo Liso/patología , Proteínas RGS/inmunología , Hipersensibilidad Respiratoria/patología , Alérgenos/administración & dosificación , Animales , Aspergillus fumigatus/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Señalización del Calcio , Femenino , Regulación de la Expresión Génica , Inyecciones Intraperitoneales , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-33 , Interleucinas/genética , Interleucinas/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ratones , Ratones Noqueados , Mucinas/inmunología , Contracción Muscular , Miocitos del Músculo Liso/inmunología , Pletismografía , Proteínas RGS/deficiencia , Proteínas RGS/genética , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Mucosa Respiratoria , Técnicas de Cultivo de Tejidos
18.
Immunogenetics ; 65(2): 145-56, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23129146

RESUMEN

Regulators of G protein signaling (RGS) are key regulators of G protein signaling. RGS proteins of the R4 RGS group are composed of a mere RGS domain and are mainly involved in immune response modulation. In both human and mouse, most genes encoding the R4 RGS proteins are located in the same region of chromosome 1. We show here that the RGS1/RGS16 neighborhood constitutes a synteny group well conserved across tetrapods and closely linked to the MHC paralogon of chromosome 1. Genes located in the RGS1/RGS16 region have paralogs close to the MHC on chromosome 6 or close to the other MHC paralogons. In amphioxus, a cephalochordate, these genes possess orthologs that are located in the same scaffolds as a number of markers defining the proto-MHC in this species (Abi-Rached et al., Nat Genet 31:100-115, 2002). We therefore propose that the RGS1/RGS16 region provides useful markers to investigate the origins and the evolution of the MHC. In addition, we show that some genes of the region appear to have immune functions not only in human, but also in Xenopus.


Asunto(s)
Complejo Mayor de Histocompatibilidad/genética , Proteínas RGS/genética , Sintenía , Animales , Evolución Molecular , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Orden Génico , Humanos , Interferones/farmacología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Mapeo Físico de Cromosoma , Proteínas RGS/inmunología , Vertebrados
19.
Mol Cell Biol ; 32(22): 4561-71, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22966200

RESUMEN

Neutrophils are first responders rapidly mobilized to inflammatory sites by a tightly regulated, nonredundant hierarchy of chemoattractants. These chemoattractants engage neutrophil cell surface receptors triggering heterotrimeric G-protein Gα(i) subunits to exchange GDP for GTP. By limiting the duration that Gα(i) subunits remain GTP bound, RGS proteins modulate chemoattractant receptor signaling. Here, we show that neutrophils with a genomic knock in of a mutation that disables regulator of G-protein signaling (RGS)-Gα(i2) interactions accumulate in the bone marrow and mobilize poorly to inflammatory sites. These defects are attributable to enhanced sensitivity to background signals, prolonged chemoattractant receptor signaling, and inappropriate CXCR2 downregulation. Intravital imaging revealed a failure of the mutant neutrophils to accumulate at and stabilize sites of sterile inflammation. Furthermore, these mice could not control a nonlethal Staphylococcus aureus infection. Neutrophil RGS proteins establish a threshold for Gα(i) activation, helping to coordinate desensitization mechanisms. Their loss renders neutrophils functionally incompetent.


Asunto(s)
Quimiotaxis/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Inflamación/enzimología , Neutrófilos/metabolismo , Proteínas RGS/metabolismo , Infecciones Estafilocócicas/enzimología , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Quimiotaxis/inmunología , Regulación hacia Abajo , Activación Enzimática , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Técnicas de Sustitución del Gen , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Inflamación/complicaciones , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Transgénicos , Mutación , Neutrófilos/inmunología , Neutrófilos/patología , Proteínas RGS/genética , Proteínas RGS/inmunología , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Factores de Tiempo
20.
J Allergy Clin Immunol ; 130(4): 968-76.e3, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22704538

RESUMEN

BACKGROUND: Drugs targeting individual G protein-coupled receptors are used as asthma therapies, but this strategy is limited because of G protein-coupled receptor signal redundancy. Regulator of G protein signaling 2 (RGS2), an intracellular selective inhibitor of multiple bronchoconstrictor receptors, may play a central role in the pathophysiology and treatment of asthma. OBJECTIVE: We defined functions and mechanisms of RGS2 in regulating airway hyperresponsiveness (AHR), the pathophysiologic hallmark of asthma. METHODS: Real-time PCR and Western blot were used to determine changes in RGS2 expression in ovalbumin-sensitized/-challenged mice. We also used immunohistochemistry and real-time PCR to compare RGS2 expression between human asthmatic and control subjects. The AHR of RGS2 knockout mice was assessed by using invasive tracheostomy and unrestrained plethysmography. Effects of loss of RGS2 on mouse airway smooth muscle (ASM) remodeling, contraction, intracellular Ca(2+), and mitogenic signaling were determined in vivo and in vitro. RESULTS: RGS2 was highly expressed in human and murine bronchial epithelium and ASM and was markedly downregulated in lungs of ovalbumin-sensitized/-challenged mice. Lung tissues and blood monocytes from asthma patients expressed significantly lower RGS2 protein (lung) and mRNA (monocytes) than from nonasthma subjects. The extent of reduction of RGS2 on human monocytes correlated with increased AHR. RGS2 knockout caused spontaneous AHR in mice. Loss of RGS2 augmented Ca(2+) mobilization and contraction of ASM cells. Loss of RGS2 also increased ASM mass and stimulated ASM cell growth via extracellular signal-regulated kinase and phosphatidylinositol 3-kinase pathways. CONCLUSION: We identified RGS2 as a potent modulator of AHR and a potential novel therapeutic target for asthma.


Asunto(s)
Hiperreactividad Bronquial/etiología , Proteínas RGS/inmunología , Proteínas RGS/fisiología , Animales , Calcio/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas RGS/antagonistas & inhibidores , Proteínas RGS/deficiencia , Proteínas RGS/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA