Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 909932, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591220

RESUMO

Introduction: Tumor microenvironment (TME) has been shown to be extensively involved in tumor development. However, the dynamic change of TME components and their effects are still unclear. Here, we attempted to identify TME-related genes that could help predict survival and may be potential therapeutic targets. Methods: Data was collected from UCSC Xena and GEO database. ESTIMATE and CIBERSORT algorithms were applied to estimate the components and the proportions of TIICs in TME. We analyzed the gene expression differences of immune components and stromal components, respectively, and finally got the overlapped DEGs. Through protein-protein interaction (PPI) network and univariate Cox regression analysis based on shared DEGs, we screened out and validated the TME-related genes. Focusing on this gene, we analyzed the expression and prognostic value of this gene, and investigated its relationship with immune cells by correlation analysis, single cell analysis, immunohistochemistry and immunofluorescence analysis. Results: Through a series analysis, we found that the proportion of immune and stromal components was an important prognostic factor, and screened out a key gene, LPAR5, which was highly correlated with prognosis and metastasis. And the expression of LPAR5 was positively correlated with immune cells, especially macrophages, indicating LPAR5+ macrophages played an important role in tumor microenvironment of osteosarcoma. Meanwhile, the genes in LPAR5 high expression group were enriched in immune-related activities and pathways, and differentially expressed genes between LPAR5+ macrophages and LPAR5- macrophages were enriched in the biological processes associated with phagocytosis and antigen presentation. What' more, we found that LPAR5 was mainly expressed in TME, and high LPAR5 expression predicting a better prognosis. Conclusion: We identified a TME-related gene, LPAR5, which is a promising indicator for TME remodeling in osteosarcoma. Particularly, LPAR5+ macrophages might have great potential to be a prognostic factor and therapeutic target for osteosarcoma.


Assuntos
Neoplasias Ósseas , Macrófagos , Osteossarcoma , Receptores de Ácidos Lisofosfatídicos , Microambiente Tumoral , Humanos , Neoplasias Ósseas/genética , Neoplasias Ósseas/imunologia , Macrófagos/imunologia , Osteossarcoma/genética , Osteossarcoma/imunologia , Fagocitose/genética , Fagocitose/imunologia , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/imunologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Prognóstico , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia
2.
J Neuroinflammation ; 18(1): 293, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34920725

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) is a pleiotropic lipid messenger that addresses at least six specific G-protein coupled receptors. Accumulating evidence indicates a significant involvement of LPA in immune cell regulation as well as Schwann cell physiology, with potential relevance for the pathophysiology of peripheral neuroinflammation. However, the role of LPA signaling in inflammatory neuropathies has remained completely undefined. Given the broad expression of LPA receptors on both Schwann cells and cells of the innate and adaptive immune system, we hypothesized that inhibition of LPA signaling may ameliorate the course of disease in experimental autoimmune neuritis (EAN). METHODS: We induced active EAN by inoculation of myelin protein 2 peptide (P255-78) in female Lewis rats. Animals received the orally available LPA receptor antagonist AM095, specifically targeting the LPA1 receptor subtype. AM095 was administered daily via oral gavage in a therapeutic regimen from 10 until 28 days post-immunization (dpi). Analyses were based on clinical testing, hemogram profiles, immunohistochemistry and morphometric assessment of myelination. RESULTS: Lewis rats treated with AM095 displayed a significant improvement in clinical scores, most notably during the remission phase. Cellular infiltration of sciatic nerve was only discretely affected by AM095. Hemogram profiles indicated no impact on circulating leukocytes. However, sciatic nerve immunohistochemistry revealed a reduction in the number of Schwann cells expressing the dedifferentiation marker Sox2 paralleled by a corresponding increase in differentiating Sox10-positive Schwann cells. In line with this, morphometric analysis of sciatic nerve semi-thin sections identified a significant increase in large-caliber myelinated axons at 28 dpi. Myelin thickness was unaffected by AM095. CONCLUSION: Thus, LPA1 signaling may present a novel therapeutic target for the treatment of inflammatory neuropathies, potentially affecting regenerative responses in the peripheral nerve by modulating Schwann cell differentiation.


Assuntos
Desdiferenciação Celular/fisiologia , Neurite Autoimune Experimental/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Células de Schwann/imunologia , Transdução de Sinais/fisiologia , Animais , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Desdiferenciação Celular/efeitos dos fármacos , Feminino , Isoxazóis/farmacologia , Isoxazóis/uso terapêutico , Neurite Autoimune Experimental/tratamento farmacológico , Neurite Autoimune Experimental/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
EBioMedicine ; 67: 103372, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33993055

RESUMO

BACKGROUND: GPR87 is a G-protein receptor that is specifically expressed in tumour cells, such as lung cancer, and rarely expressed in normal cells. GPR87 is a promising target for cancer therapy, but its ligand is controversial. Near-infrared photoimmunotherapy (NIR-PIT) is a novel cancer therapy in which a photosensitiser, IRDye700DX (IR700), binds to antibodies and specifically destroys target cells by irradiating them with near-infrared-light. Here, we aimed to develop a NIR-PIT targeting GPR87. METHODS: We evaluated the expression of GPR87 in resected specimens of lung cancer and malignant pleural mesothelioma (MPM) resected at Nagoya University Hospital using immunostaining. Humanised anti-GPR87 antibody (huGPR87) was generated by introducing CDRs from mouse anti-GPR87 antibody generated by standard hybridoma method. HuGPR87 was conjugated with IR700 and the therapeutic effect of NIR-PIT was evaluated in vitro and in vivo using lung cancer or MPM cell lines. FINDINGS: Among the surgical specimens, 54% of lung cancer and 100% of MPM showed high expression of GPR87. It showed therapeutic effects on lung cancer and MPM cell lines in vitro, and showed therapeutic effects in multiple models in vivo. INTERPRETATION: These results suggest that NIR-PIT targeting GPR87 is a promising therapeutic approach for the treatment of thoracic cancer. FUNDING: This research was supported by the Program for Developing Next-generation Researchers (Japan Science and Technology Agency), KAKEN (18K15923, 21K07217, JSPS), FOREST-Souhatsu, CREST (JST).


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Imunoterapia/métodos , Neoplasias Pulmonares/terapia , Fototerapia/métodos , Receptores de Ácidos Lisofosfatídicos/imunologia , Células 3T3 , Animais , Anticorpos Monoclonais Humanizados/imunologia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Feminino , Humanos , Raios Infravermelhos , Masculino , Camundongos , Camundongos Nus
5.
Front Immunol ; 11: 531910, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584637

RESUMO

Immunological tolerance has evolved to curtail immune responses against self-antigens and prevent autoimmunity. One mechanism that contributes to immunological tolerance is the expression of inhibitory receptors by lymphocytes that signal to dampen immune responses during the course of an infection and to prevent immune-mediated collateral damage to the host. The understanding that tumors exploit these physiological mechanisms to avoid elimination has led to remarkable, but limited, success in the treatment of cancer through the use of biologics that interfere with the ability of cancers to suppress immune function. This therapy, based on the understanding of how T lymphocytes are normally activated and suppressed, has led to the development of therapeutic blocking antibodies, referred to as immune checkpoint blockade, which either directly or indirectly promote the activation of CD8 T cells to eradicate cancer. Here, we highlight the distinct signaling mechanisms, timing and location of inhibition used by the CTLA-4 and PD-1 inhibitory receptors compared to a novel inhibitory signaling axis comprised of the bioactive lipid, lysophosphatidic acid (LPA), signaling via the LPA5 receptor expressed by CD8 T cells. Importantly, abundant evidence indicates that an LPA-LPA5 signaling axis is also exploited by diverse cancers to suppress T cell activation and function. Clearly, a thorough molecular and biochemical understanding of how diverse T cell inhibitory receptors signal to suppress T cell antigen receptor signaling and function will be important to inform the choice of which complimentary checkpoint blockade modalities might be used for a given cancer.


Assuntos
Antígeno CTLA-4/imunologia , Lisofosfolipídeos/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Evasão Tumoral , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Neoplasias/patologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Transdução de Sinais/imunologia
6.
Front Immunol ; 10: 1159, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231367

RESUMO

Persistent T cell antigen receptor (TCR) signaling by CD8 T cells is a feature of cancer and chronic infections and results in the sustained expression of, and signaling by, inhibitory receptors, which ultimately impair cytotoxic activity via poorly characterized mechanisms. We have previously determined that the LPA5 GPCR expressed by CD8 T cells, upon engaging the lysophosphatidic acid (LPA) bioactive serum lipid, functions as an inhibitory receptor able to negatively regulate TCR signaling. Notably, the levels of LPA and autotaxin (ATX), the phospholipase D enzyme that produces LPA, are often increased in chronic inflammatory disorders such as chronic infections, autoimmune diseases, obesity, and cancer. In this report, we demonstrate that LPA engagement selectively by LPA5 on human and mouse CD8 T cells leads to the inhibition of several early TCR signaling events including intracellular calcium mobilization and ERK activation. We further show that, as a consequence of LPA5 suppression of TCR signaling, the exocytosis of perforin-containing granules is significantly impaired and reflected by repressed in vitro and in vivo CD8 T cell cytolytic activity. Thus, these data not only document LPA5 as a novel inhibitory receptor but also determine the molecular and biochemical mechanisms by which a naturally occurring serum lipid that is elevated under settings of chronic inflammation signals to suppress CD8 T cell killing activity in both human and murine cells. As diverse tumors have repeatedly been shown to aberrantly produce LPA that acts in an autocrine manner to promote tumorigenesis, our findings further implicate LPA in activating a novel inhibitory receptor whose signaling may be therapeutically silenced to promote CD8 T cell immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Cálcio/imunologia , Cálcio/metabolismo , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Exocitose/imunologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perforina/imunologia , Perforina/metabolismo , Diester Fosfórico Hidrolases/imunologia , Diester Fosfórico Hidrolases/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/genética , Linfócitos T Citotóxicos/metabolismo
7.
Am J Respir Cell Mol Biol ; 54(3): 402-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26248018

RESUMO

Lysophosphatidic acid (LPA) is a pleiotropic lipid signaling molecule associated with asthma pathobiology. LPA elicits its effects by binding to at least six known cell surface G protein-coupled receptors (LPA1-6) that are expressed in the lung in a cell type-specific manner. LPA2 in particular has emerged as an attractive therapeutic target in asthma because it appears to transduce inhibitory or cell-protective signals. We studied a novel and specific small molecule LPA2 agonist (2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl] benzoic acid [DBIBB]) in a mouse model of house dust mite-induced allergic airway inflammation. Mice injected with DBIBB developed significantly less airway and lung inflammation compared with vehicle-treated controls. Levels of lung Th2 cytokines were also significantly attenuated by DBIBB. We conclude that pharmacologic activation of LPA2 attenuates Th2-driven allergic airway inflammation in a mouse model of asthma. Targeting LPA receptor signaling holds therapeutic promise in allergic asthma.


Assuntos
Antiasmáticos/farmacologia , Anti-Inflamatórios/farmacologia , Asma/prevenção & controle , Pulmão/efeitos dos fármacos , Naftalimidas/farmacologia , Pneumonia/prevenção & controle , Receptores de Ácidos Lisofosfatídicos/agonistas , Sulfonamidas/farmacologia , Alérgenos , Animais , Antígenos de Dermatophagoides , Proteínas de Artrópodes , Asma/imunologia , Asma/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Camundongos Endogâmicos BALB C , Diester Fosfórico Hidrolases/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Receptores de Ácidos Lisofosfatídicos/imunologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo , Fatores de Tempo
8.
J Immunol ; 195(3): 1139-51, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26101324

RESUMO

Blood-derived monocytes remove apoptotic cells and terminate inflammation in settings as diverse as atherosclerosis and Alzheimer's disease. They express high levels of the proresolving receptor ALX/FPR2, which is activated by the protein annexin A1 (ANXA1), found in high abundance in inflammatory exudates. Using primary human blood monocytes from healthy donors, we identified ANXA1 as a potent CD14(+)CD16(-) monocyte chemoattractant, acting via ALX/FPR2. Downstream signaling pathway analysis revealed the p38 MAPK-mediated activation of a calcium independent phospholipase A2 with resultant synthesis of lysophosphatidic acid (LPA) driving chemotaxis through LPA receptor 2 and actin cytoskeletal mobilization. In vivo experiments confirmed ANXA1 as an independent phospholipase A2-dependent monocyte recruiter; congruently, monocyte recruitment was significantly impaired during ongoing zymosan-induced inflammation in AnxA1(-/-) or alx/fpr2/3(-/-) mice. Using a dorsal air-pouch model, passive transfer of apoptotic neutrophils between AnxA1(-/-) and wild-type mice identified effete neutrophils as the primary source of soluble ANXA1 in inflammatory resolution. Together, these data elucidate a novel proresolving network centered on ANXA1 and LPA generation and identify previously unappreciated determinants of ANXA1 and ALX/FPR2 signaling in monocytes.


Assuntos
Anexina A1/imunologia , Apoptose/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Citoesqueleto de Actina/metabolismo , Animais , Anexina A1/genética , Células Cultivadas , Ativação Enzimática/imunologia , Humanos , Inflamação/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Lisofosfolipídeos/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/transplante , Fosfolipases A2 Independentes de Cálcio/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Formil Peptídeo/biossíntese , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de IgG/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Zimosan , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
J Immunol ; 193(2): 617-26, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24935929

RESUMO

T cells exhibit high-speed migration within the paracortical T zone of lymph nodes (LNs) as they scan cognate Ags displayed by dendritic cells in the tissue microenvironment supported by the network of stromal cells. Although intranodal T cell migration is controlled in part by chemokines and LFA-1/ICAM-1, the mechanisms underlying their migratory activity independent of these factors remain to be elucidated. In this study, we show that LN stromal cells constitutively express autotaxin (ATX), an ectoenzyme that is important for the generation of lysophosphatidic acid (LPA). Importantly, CCL21(+) stromal cells in the T zone produced and immobilized ATX on their cell surface. Two-photon imaging using LN tissue slices revealed that pharmacological inhibition of ATX or LPA receptors significantly reduced T cell migration, and this was further exacerbated by blockage of Gαi signaling or LFA-1. Therefore, T cell motility mediated by the ATX-LPA axis was independent of Gαi and LFA-1. LPA induced slow intermittent movement of T cells in vitro in a LFA-1-independent manner and enhanced CCL21-induced migration. Moreover, LPA and CCL21 cooperatively augmented RhoA activity in T cells, which was necessary for efficient intranodal T cell migration via the downstream ROCK-myosin II pathway. Taken together, T zone stromal cells control optimal migratory behavior of T cells via multiple signaling cues mediated by chemokines and ATX/LPA.


Assuntos
Movimento Celular/imunologia , Linfonodos/imunologia , Diester Fosfórico Hidrolases/imunologia , Células Estromais/imunologia , Linfócitos T/imunologia , Proteína rhoA de Ligação ao GTP/imunologia , Anilidas/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Quimiocina CCL21/farmacologia , Feminino , Isoxazóis/farmacologia , Linfonodos/metabolismo , Antígeno-1 Associado à Função Linfocitária/genética , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência por Excitação Multifotônica , Miosina Tipo II/genética , Miosina Tipo II/imunologia , Miosina Tipo II/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Organofosfonatos/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Propionatos/farmacologia , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/imunologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Estromais/metabolismo , Linfócitos T/citologia , Transcriptoma/genética , Transcriptoma/imunologia , Quinases Associadas a rho/genética , Quinases Associadas a rho/imunologia , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
10.
J Immunol ; 193(1): 85-95, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24890721

RESUMO

Lysophospholipids have emerged as biologically important chemoattractants capable of directing lymphocyte development, trafficking, and localization. Lysophosphatidic acid (LPA) is a major lysophospholipid found systemically, and its levels are elevated in certain pathological settings, such as cancer and infections. In this study, we demonstrate that BCR signal transduction by mature murine B cells is inhibited upon LPA engagement of the LPA5 (GPR92) receptor via a Gα12/13-Arhgef1 pathway. The inhibition of BCR signaling by LPA5 manifests by impaired intracellular calcium store release and most likely by interfering with inositol 1,4,5-triphosphate receptor activity. We further show that LPA5 also limits Ag-specific induction of CD69 and CD86 expression and that LPA5-deficient B cells display enhanced Ab responses. Thus, these data show that LPA5 negatively regulates BCR signaling, B cell activation, and immune response. Our findings extend the influence of lysophospholipids on immune function and suggest that alterations in LPA levels likely influence adaptive humoral immunity.


Assuntos
Formação de Anticorpos/fisiologia , Linfócitos B/imunologia , Imunidade Humoral/fisiologia , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Transdução de Sinais/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígeno B7-2/genética , Antígeno B7-2/imunologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos B/genética , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais/genética
11.
J Immunol ; 192(3): 851-7, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24443508

RESUMO

Lysophosphatidic acid (LPA) is a pleiotropic lipid molecule with potent effects on cell growth and motility. Major progress has been made in recent years in deciphering the mechanisms of LPA generation and how it acts on target cells. Most research has been conducted in other disciplines, but emerging data indicate that LPA has an important role to play in immunity. A key discovery was that autotaxin (ATX), an enzyme previously implicated in cancer cell motility, generates extracellular LPA from the precursor lysophosphatidylcholine. Steady-state ATX is expressed by only a few tissues, including high endothelial venules in lymph nodes, but inflammatory signals can upregulate ATX expression in different tissues. In this article, we review current thinking about the ATX/LPA axis in lymphocyte homing, as well as in models of allergic airway inflammation and asthma. New insights into the role of LPA in regulating immune responses should be forthcoming in the near future.


Assuntos
Quimiotaxia de Leucócito/imunologia , Lisofosfolipídeos/imunologia , Diester Fosfórico Hidrolases/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Remodelação das Vias Aéreas/imunologia , Animais , Antígenos/administração & dosagem , Antígenos/imunologia , Asma/imunologia , Movimento Celular/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Inflamação/imunologia , Isoxazóis/farmacologia , Tecido Linfoide/imunologia , Lisofosfolipídeos/análise , Camundongos , Diester Fosfórico Hidrolases/efeitos dos fármacos , Propionatos/farmacologia , Quimera por Radiação , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/imunologia , Transdução de Sinais/imunologia , Regulação para Cima , Vênulas/imunologia
12.
J Immunol ; 188(8): 3784-90, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22427635

RESUMO

Negative regulation of innate immune responses is essential to prevent excess inflammation and tissue injury and promote homeostasis. Lysophosphatidic acid (LPA) is a pleiotropic lipid that regulates cell growth, migration, and activation and is constitutively produced at low levels in tissues and in serum. Extracellular LPA binds to specific G protein-coupled receptors, whose function in regulating innate or adaptive immune responses remains poorly understood. Of the classical LPA receptors belonging to the Edg family, lpa2 (edg4) is expressed by dendritic cells (DC) and other innate immune cells. In this article, we show that DC from lpa2(-/-) mice are hyperactive compared with their wild-type counterparts and are less susceptible to inhibition by different LPA species. In transient-transfection assays, we found that lpa2 overexpression inhibits NF-κB-driven gene transcription. Using an adoptive-transfer approach, we found that allergen-pulsed lpa2(-/-) DC induced substantially more lung inflammation than did wild-type DC after inhaled allergen challenge. Finally, lpa2(-/-) mice develop greater allergen-driven lung inflammation than do their wild-type counterparts in models of allergic asthma involving both systemic and mucosal sensitization. Taken together, these findings identify LPA acting via lpa2 as a novel negative regulatory pathway that inhibits DC activation and allergic airway inflammation.


Assuntos
Asma/imunologia , Células Dendríticas/imunologia , Pulmão/imunologia , Lisofosfolipídeos/imunologia , NF-kappa B/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Administração por Inalação , Transferência Adotiva , Alérgenos/imunologia , Animais , Asma/patologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células HEK293 , Humanos , Inflamação/imunologia , Inflamação/patologia , Pulmão/metabolismo , Pulmão/patologia , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , Receptores de Ácidos Lisofosfatídicos/deficiência , Receptores de Ácidos Lisofosfatídicos/genética , Transdução de Sinais , Transcrição Gênica
13.
Int Immunol ; 21(6): 667-77, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19461126

RESUMO

Lysophosphatidic acid (LPA) is an activator and chemoattractant of NK cells, which are critical members of the immunological tumor surveillance machinery. Here, we analyzed the influence of LPA on the interaction of human NK cells with tumor cells such as the Burkitt lymphoma cell line Raji and the human melanoma cell line A2058. Thereby we found that LPA inhibits the release of perforin and cytotoxic activity of NK cells. Analysis of signal transduction showed that LPA induces common signaling pathways of chemotaxins such as G(i) protein-dependent actin re-organization, activation of the mitogen-activated protein kinase p38 as well as phosphatidylinositol-3-kinase-dependent signal molecules [protein kinase B/Akt and glycogen synthase kinase-3beta (GSK-3beta)]. In contrast to most chemotaxins, LPA is also able to activate G(s)-dependent signaling molecules. This signaling cascade involves the LPA receptor type-2, increase cAMP levels and protein kinase A (PKA) activation, which in turn are responsible for the modulatory effect of LPA on NK cell-mediated cytotoxicity. Moreover, blocking the regulatory subunits of PKA I abrogates the inhibitory effect of LPA, whereas the catalytic subunits are not involved. Based on our data, one can assume that LPA contributes to the tumor escape from the immunological surveillance machinery.


Assuntos
Linfoma de Burkitt/imunologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células Matadoras Naturais/metabolismo , Lisofosfolipídeos/metabolismo , Melanoma/imunologia , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patologia , Linhagem Celular Tumoral , Classe Ia de Fosfatidilinositol 3-Quinase , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citotoxicidade Imunológica , Regulação para Baixo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/imunologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/imunologia , Humanos , Vigilância Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Lisofosfolipídeos/imunologia , Melanoma/metabolismo , Melanoma/patologia , Perforina/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Ácidos Lisofosfatídicos/imunologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/imunologia , Evasão Tumoral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Immunol Res ; 45(2-3): 229-38, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19184538

RESUMO

Lysophosphatidic acid (LPA) can affect the growth, migration, and activation of many different cell types. Research in this field has recently accelerated due to the molecular cloning of LPA receptors as well as advances in our understanding of LPA metabolism. A major pathway for LPA generation is the hydrolysis of lysophosphatidylcholine by the enzyme autotaxin (ATX). Although most research to-date has been conducted in other disciplines (e.g., neurobiology and cardiovascular diseases), emerging data point to an important role for LPA and ATX in regulating immune responses. Here we review current understanding of LPA and ATX in immunity with an emphasis on migration and activation of lymphocytes and dendritic cells. New gene-targeted and transgenic mice, receptor-specific antibodies, and pathway antagonists should rapidly enhance our understanding of this versatile lysolipid in immune responses in the near future.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Lisofosfolipídeos/imunologia , Diester Fosfórico Hidrolases/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Diester Fosfórico Hidrolases/metabolismo , Receptores de Ácidos Lisofosfatídicos/imunologia , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/imunologia
15.
Breast Cancer Res ; 6(6): R640-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15535846

RESUMO

INTRODUCTION: Lysophosphatidic acid (LPA) is a bioactive phospholipid with diverse effects on various cells. It interacts with at least three G-protein-coupled transmembrane receptors, namely LPA1, LPA2 and LPA3, whose expression in various tumours has not been fully characterized. In the present study we characterized the expression profile of LPA receptors in human breast cancer tissue and assessed the possible roles of each receptor. METHODS: The relative expression levels of each receptor's mRNA against beta-actin mRNA was examined in surgically resected invasive ductal carcinomas and normal gland tissue using real-time RT-PCR. LPA2 expression was also examined immunohistochemically using a rat anti-LPA2 monoclonal antibody. RESULTS: In 25 cases normal and cancer tissue contained LPA1 mRNA at similar levels, whereas the expression level of LPA2 mRNA was significantly increased in cancer tissue as compared with its normal counterpart (3479.0 +/- 426.6 versus 1287.3 +/- 466.8; P < 0.05). LPA3 was weakly expressed in both cancer and normal gland tissue. In 48 (57%) out of 84 cases, enhanced expression of LPA2 protein was confirmed in carcinoma cells as compared with normal mammary epithelium by immunohistochemistry. Over-expression of LPA2 was detected in 17 (45%) out of 38 premenopausal women, as compared with 31 (67%) out of 46 postmenopausal women, and the difference was statistically significant (P < 0.05). CONCLUSION: These findings suggest that upregulation of LPA2 may play a role in carcinogenesis, particularly in postmenopausal breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Receptores de Ácidos Lisofosfatídicos/biossíntese , Anticorpos Monoclonais/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Blood ; 104(13): 4080-7, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15319282

RESUMO

Mast cells (MCs) initiate immune responses from mucosal surfaces and perivascular spaces. Stem cell factor (SCF) regulates MC development and viability, but the role of innate serum factors in MC development is unexplored. Cultured cord blood-derived human MCs (hMCs) express mRNA transcripts for all 4 known receptors for lysophosphatidic acid (LPA), an abundant serum-associated lipid growth factor. In an SCF-dependent serum-free culture system, LPA (2.5-10 microM) increased the total number of hMCs by approximately 10-fold compared with cultures maintained in the absence of LPA under otherwise identical conditions. LPA was comitogenic with SCF but did not prolong MC survival. LPA-mediated proliferation was blocked by VPC-32179, a competitive antagonist of LPA(1) and LPA(3) receptors, and by pertussis toxin, and it was also attenuated by GW9662, a selective antagonist of peroxisome proliferator-activated receptor (PPAR)-gamma. LPA accelerated the acquisition of hMC granules and increased Kit expression. hMCs derived in the presence of LPA were functional, as evidenced by their immunoglobulin E (IgE)-dependent histamine release and by their characteristic proliferative responses to interleukin-3 (IL-3), IL-4, and IL-9 in combination with SCF. Thus, LPA acts through LPA receptor and PPAR-gamma-dependent pathways to accelerate hMC proliferation and differentiation, and it modulates their phenotype without providing cytoprotection. LPA could facilitate MC hyperplasia in inflammation associated with either innate or adaptive immunity.


Assuntos
Lisofosfolipídeos/farmacologia , Mastócitos/citologia , Apoptose , Sequência de Bases , Divisão Celular , Primers do DNA , Sangue Fetal , Humanos , Recém-Nascido , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , RNA Mensageiro/genética , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/imunologia , Cordão Umbilical
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA