Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 630
Filtrar
1.
J Immunol ; 208(8): 1912-1923, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35379745

RESUMO

The mechanism regulating the life span of short-lived plasma cells (SLPCs) remains poorly understood. Here we demonstrated that the EP4-mediated activation of AKT by PGE2 was required for the proper control of inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) hyperactivation and hence the endoplasmic reticulum (ER) homeostasis in IgM-producing SLPCs. Disruption of the PGE2-EP4-AKT signaling pathway resulted in IRE1α-induced activation of JNK, leading to accelerated death of SLPCs. Consequently, Ptger4-deficient mice (C57BL/6) exhibited a markedly impaired IgM response to T-independent Ags and increased susceptibility to Streptococcus pneumoniae infection. This study reveals a highly selective impact of the PGE2-EP4 signal on the humoral immunity and provides a link between ER stress response and the life span of SLPCs.


Assuntos
Sobrevivência Celular , Dinoprostona , Estresse do Retículo Endoplasmático , Endorribonucleases , Plasmócitos , Proteínas Serina-Treonina Quinases , Animais , Sobrevivência Celular/imunologia , Dinoprostona/imunologia , Estresse do Retículo Endoplasmático/imunologia , Endorribonucleases/imunologia , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Prostaglandinas/imunologia , Prostaglandinas E/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia
2.
Biomed J ; 45(1): 109-117, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34175493

RESUMO

Leishmaniasis is a neglected tropical disease that causes several clinical manifestations. Parasites of the genus Leishmania cause this disease. Spread across five continents, leishmaniasis is a particular public health problem in developing countries. Leishmania infects phagocytic cells such as macrophages, where it induces adenosine triphosphate (ATP) release at the time of infection. ATP activates purinergic receptors in the cell membranes of infected cells and promotes parasite control by inducing leukotriene B4 release and NLRP3 inflammasome activation. Moreover, uridine triphosphate induces ATP release, exacerbating the immune response. However, ATP may also undergo catalysis by ectonucleotidases present in the parasite membrane, generating adenosine, which activates P1 receptors and induces the production of anti-inflammatory molecules such as prostaglandin E2 and IL-10. These mechanisms culminate in Leishmania's survival. Thus, how Leishmania handles extracellular nucleotides and the activation of purinergic receptors determines the control or the dissemination of the disease.


Assuntos
Leishmania , Leishmaniose , Receptores Purinérgicos , Adenosina , Trifosfato de Adenosina , Dinoprostona/imunologia , Humanos , Interleucina-10/imunologia , Leishmania/fisiologia , Leishmaniose/imunologia , Leucotrieno B4/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Receptores Purinérgicos/metabolismo , Transdução de Sinais
3.
Aging (Albany NY) ; 13(18): 22134-22147, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34550907

RESUMO

BACKGROUND: Osteoarthritis (OA) is degenerative joint disorder mainly characterized by long-term pain with limited activity of joints, the disease has no effective preventative therapy. Rutin (RUT) is a flavonoid compound, present naturally. The flavonoid shows range of biological activities such as anti-inflammatory and anti-cancer effect. We screened RUT for its activity against osteoarthritis with in vivo and in vitro models of osteoarthritis. METHODS: Animal model of OA was developed using C57BL/6 mice by surgical destabilization of medial meniscus. For in vitro studies the human articular cartilage tissues were used which were collected from osteoarthritis patients and were processed to isolate chondrocytes. The chondrocytes were submitted to advanced glycation end products (AGEs) for inducing osteoarthritis in vitro. Cell viability was done by CCK-8 assay, ELISA analysis for MMP13, collage II, PGE2, IL-6, TNF-α, ADAMTS-5 and MMP-13. Western blot analysis was done for expression of proteins and in silico analysis was done by docking studies. RESULTS: Pretreatment of RT showed no cytotoxic effect and also ameliorated the AGE mediated inflammatory reaction on human chondrocytes in vitro. Treatment of RT inhibited the levels of COX-2 and iNOS in AGE exposed chondrocytes. RT decreased the AGE mediated up-regulation of IL-6, NO, TNF-α and PGE-2 in a dose dependent manner. Pretreatment of RT decreased the extracellular matrix degradation, inhibited expression of TRAF-6 and BCL-2 the NF-κB/MAPK pathway proteins. The treatment of RT in mice prevented the calcification of cartilage tissues, loss of proteoglycans and also halted the narrowing of joint space is mice subjected to osteoarthritis. The in-silico analysis suggested potential binding affinity of RT with TRAF-6 and BCL-2. CONCLUSION: In brief RT inhibited AGE-induced inflammatory reaction and also degradation of ECM via targeting the NF-κB/MAPK pathway proteins BCL-2 and TRAF-6. RT can be a potential molecule in treating OA.


Assuntos
Anti-Inflamatórios/administração & dosagem , Matriz Extracelular/imunologia , Produtos Finais de Glicação Avançada/imunologia , Osteoartrite/tratamento farmacológico , Osteoartrite/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Rutina/administração & dosagem , Fator 6 Associado a Receptor de TNF/imunologia , Animais , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Condrócitos/efeitos dos fármacos , Condrócitos/imunologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/imunologia , Osteoartrite/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator 6 Associado a Receptor de TNF/genética
4.
PLoS Pathog ; 17(9): e1009493, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34555127

RESUMO

Listeria monocytogenes is an intracellular bacterium that elicits robust CD8+ T-cell responses. Despite the ongoing development of L. monocytogenes-based platforms as cancer vaccines, our understanding of how L. monocytogenes drives robust CD8+ T-cell responses remains incomplete. One overarching hypothesis is that activation of cytosolic innate pathways is critical for immunity, as strains of L. monocytogenes that are unable to access the cytosol fail to elicit robust CD8+ T-cell responses and in fact inhibit optimal T-cell priming. Counterintuitively, however, activation of known cytosolic pathways, such as the inflammasome and type I IFN, lead to impaired immunity. Conversely, production of prostaglandin E2 (PGE2) downstream of cyclooxygenase-2 (COX-2) is essential for optimal L. monocytogenes T-cell priming. Here, we demonstrate that vacuole-constrained L. monocytogenes elicit reduced PGE2 production compared to wild-type strains in macrophages and dendritic cells ex vivo. In vivo, infection with wild-type L. monocytogenes leads to 10-fold increases in PGE2 production early during infection whereas vacuole-constrained strains fail to induce PGE2 over mock-immunized controls. Mice deficient in COX-2 specifically in Lyz2+ or CD11c+ cells produce less PGE2, suggesting these cell subsets contribute to PGE2 levels in vivo, while depletion of phagocytes with clodronate abolishes PGE2 production completely. Taken together, this work demonstrates that optimal PGE2 production by phagocytes depends on L. monocytogenes access to the cytosol, suggesting that one reason cytosolic access is required to prime CD8+ T-cell responses may be to facilitate production of PGE2.


Assuntos
Células Dendríticas/imunologia , Dinoprostona/biossíntese , Dinoprostona/imunologia , Listeriose/imunologia , Macrófagos/imunologia , Animais , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Feminino , Listeria monocytogenes/imunologia , Ativação Linfocitária/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Aging (Albany NY) ; 13(16): 20651-20660, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34438368

RESUMO

NOD-like receptor family caspase recruitment domain family domain containing 5 (NLRC5) has been found to be a critical mediator of inflammatory response. However, the role of NLRC5 in osteoarthritis (OA) has not been reported. Our results showed that NLRC5 was down-regulated by IL-1ß induction in chondrocytes. Overexpression of NLRC5 in chondrocytes significantly suppressed IL-1ß-induced inflammatory response through inhibiting the production of multiple inflammatory mediators including inducible nitric oxide synthases (iNOS), and cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), NO, TNF-α and IL-6, as well matrix metalloproteinase 3 (MMP-3) and MMP-13. Consistently, NLRC5 knockdown exhibited opposite effects on the production of these inflammatory mediators in IL-1ß-induced chondrocytes. Furthermore, overexpression of NLRC5 increased the IĸBα expression, while decreased the p-p65 expression, indicating that NLRC5 inhibited the activation of NF-κB signaling. Additionally, inhibition of NF-κB by PDTC mitigated the si-NLRC5-mediated promotion of IL-1ß-induced inflammatory injury in chondrocytes. Finally, NLRC5 treatment ameliorated cartilage degeneration in an OA model in rats. Taken together, these findings revealed that NLRC5 attenuated IL-1ß-induced inflammatory injury in chondrocytes through regulating the NF-κB signaling.


Assuntos
Condrócitos/imunologia , Interleucina-1beta/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , NF-kappa B/imunologia , Osteoartrite/imunologia , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , Humanos , Interleucina-1beta/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/imunologia , NF-kappa B/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Osteoartrite/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
6.
Microbiol Spectr ; 9(1): e0049621, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34319170

RESUMO

Prostaglandin E2 (PGE2) is an essential immunomodulatory lipid released by cells in response to infection with many bacteria, yet its function in macrophage-mediated bacterial clearance is poorly understood. Yersinia overall inhibits the inflammatory circuit, but its effect on PGE2 production is unknown. We hypothesized that one of the Yersinia effector proteins is responsible for the inhibition of PGE2 biosynthesis. We identified that yopB-deficient Y. enterocolitica and Y. pseudotuberculosis deficient in the secretion of virulence proteins via a type 3 secretion system (T3SS) failed to inhibit PGE2 biosynthesis in macrophages. Consistently, COX-2-mediated PGE2 biosynthesis is upregulated in cells treated with heat-killed or T3SS-deficient Y. pseudotuberculosis but diminished in the presence of a MAPK/ERK inhibitor. Mutants expressing catalytically inactive YopJ induce similar levels of PGE2 as heat-killed or ΔyopB Y. pseudotuberculosis, reversed by YopJ complementation. Shotgun proteomics discovered host pathways regulated in a YopJ-mediated manner, including pathways regulating PGE2 synthesis and oxidative phosphorylation. Consequently, this study identified that YopJ-mediated inhibition of MAPK signal transduction serves as a mechanism targeting PGE2, an alternative means of inflammasome inhibition by Yersinia. Finally, we showed that EP4 signaling supports macrophage function in clearing intracellular bacteria. In summary, our unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription, thereby enhancing the intracellular survival of yersiniae. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic molecule to improve the outcomes of specific bacterial infections. Since other pathogens encode YopJ homologs, this mechanism is expected to be present in other infections. IMPORTANCE PGE2 is a critical immunomodulatory lipid, but its role in bacterial infection and pathogen clearance is poorly understood. We previously demonstrated that PGE2 leads to macrophage polarization toward the M1 phenotype and stimulates inflammasome activation in infected macrophages. Finally, we also discovered that PGE2 improved the clearance of Y. enterocolitica. The fact that Y. enterocolitica hampers PGE2 secretion in a type 3 secretion system (T3SS)-dependent manner and because PGE2 appears to assist macrophage in the clearance of this bacterium indicates that targeting of the eicosanoid pathway by Yersinia might be an adaption used to counteract host defenses. Our study identified a mechanism used by Yersinia that obstructs PGE2 biosynthesis in human macrophages. We showed that Y. pseudotuberculosis interferes with PGE2 biosynthesis by using one of its T3SS effectors, YopJ. Specifically, YopJ targets the host COX-2 enzyme responsible for PGE2 biosynthesis, which happens in a MAPK/ER-dependent manner. Moreover, in a shotgun proteomics study, we also discovered other pathways that catalytically active YopJ targets in the infected macrophages. YopJ was revealed to play a role in limiting host LPS responses, including repression of EGR1 and JUN proteins, which control transcriptional activation of proinflammatory cytokine production such as interleukin-1ß. Since YopJ has homologs in other bacterial species, there are likely other pathogens that target and inhibit PGE2 biosynthesis. In summary, our study's unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic target.


Assuntos
Proteínas de Bactérias/imunologia , Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , MAP Quinases Reguladas por Sinal Extracelular/imunologia , Macrófagos/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Infecções por Yersinia pseudotuberculosis/microbiologia , Yersinia pseudotuberculosis/imunologia , Animais , Proteínas de Bactérias/genética , Ciclo-Oxigenase 2/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Interações Hospedeiro-Patógeno , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Transdução de Sinais , Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/imunologia
7.
Sci Rep ; 11(1): 13559, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193890

RESUMO

Prostaglandin E2 (PGE2), an active lipid compound derived from arachidonic acid, regulates different stages of the immune response of the host during several pathologies such as chronic infections or cancer. In fact, manipulation of PGE2 levels was proposed as an approach for countering the Type I IFN signature of tuberculosis (TB). However, very limited information regarding the PGE2 pathway in patients with active TB is currently available. In the present work, we demonstrated that PGE2 exerts a potent immunosuppressive action during the immune response of the human host against Mycobacterium tuberculosis (Mtb) infection. Actually, we showed that PGE2 significantly reduced the surface expression of several immunological receptors, the lymphoproliferation and the production of proinflammatory cytokines. In addition, PGE2 promoted autophagy in monocytes and neutrophils cultured with Mtb antigens. These results suggest that PGE2 might be attenuating the excessive inflammatory immune response caused by Mtb, emerging as an attractive therapeutic target. Taken together, our findings contribute to the knowledge of the role of PGE2 in the human host resistance to Mtb and highlight the potential of this lipid mediator as a tool to improve anti-TB treatment.


Assuntos
Dinoprostona/farmacologia , Imunossupressores/farmacologia , Monócitos/imunologia , Mycobacterium tuberculosis/imunologia , Neutrófilos/imunologia , Tuberculose/imunologia , Adulto , Dinoprostona/imunologia , Feminino , Humanos , Imunossupressores/imunologia , Masculino
8.
Immunity ; 54(8): 1665-1682.e14, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34129840

RESUMO

Tight control of inflammatory gene expression by antagonistic environmental cues is key to ensure immune protection while preventing tissue damage. Prostaglandin E2 (PGE2) modulates macrophage activation during homeostasis and disease, but the underlying mechanisms remain incompletely characterized. Here we dissected the genomic properties of lipopolysaccharide (LPS)-induced genes whose expression is antagonized by PGE2. The latter molecule targeted a set of inflammatory gene enhancers that, already in unstimulated macrophages, displayed poorly permissive chromatin organization and were marked by the transcription factor myocyte enhancer factor 2A (MEF2A). Deletion of MEF2A phenocopied PGE2 treatment and abolished type I interferon (IFN I) induction upon exposure to innate immune stimuli. Mechanistically, PGE2 interfered with LPS-mediated activation of ERK5, a known transcriptional partner of MEF2. This study highlights principles of plasticity and adaptation in cells exposed to a complex environment and uncovers a transcriptional circuit for IFN I induction with relevance for infectious diseases or cancer.


Assuntos
Dinoprostona/imunologia , Interferon Tipo I/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Animais , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Interferon Tipo I/biossíntese , Lipopolissacarídeos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/metabolismo
9.
Cancer Res ; 81(15): 4124-4132, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34035084

RESUMO

Prostaglandin E2 (PGE2) promotes tumor progression through evasion of antitumor immunity. In stark contrast to cyclooxygenase-dependent production of PGE2, little is known whether PGE2 secretion is regulated within tumor tissues. Here, we show that VEGF-dependent release of thromboxane A2 (TXA2) triggers Ca2+ transients in tumor cells, culminating in PGE2 secretion and subsequent immune evasion in the early stages of tumorigenesis. Ca2+ transients caused cPLA2 activation and triggered the arachidonic acid cascade. Ca2+ transients were monitored as the surrogate marker of PGE2 secretion. Intravital imaging of BrafV600E mouse melanoma cells revealed that the proportion of cells exhibiting Ca2+ transients is markedly higher in vivo than in vitro. The TXA2 receptor was indispensable for the Ca2+ transients in vivo, high intratumoral PGE2 concentration, and evasion of antitumor immunity. Notably, treatment with a VEGF receptor antagonist and an anti-VEGF antibody rapidly suppressed Ca2+ transients and reduced TXA2 and PGE2 concentrations in tumor tissues. These results identify the VEGF-TXA2 axis as a critical promoter of PGE2-dependent tumor immune evasion, providing a molecular basis underlying the immunomodulatory effect of anti-VEGF therapies. SIGNIFICANCE: This study identifies the VEGF-TXA2 axis as a potentially targetable regulator of PGE2 secretion, which provides novel strategies for prevention and treatment of multiple types of malignancies.


Assuntos
Dinoprostona/imunologia , Evasão da Resposta Imune/imunologia , Microscopia Intravital/métodos , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Humanos , Camundongos , Camundongos Nus
10.
Cell Biol Int ; 45(5): 1072-1081, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33470478

RESUMO

Mesenchymal stem cells (MSCs) play an important role as immune modulator through interaction with several immune cells, including macrophages. In this study, the immunomodulatory potency of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) was demonstrated in the in vivo middle cerebral artery occlusion (MCAo)-induced brain injury rat model and in vitro THP-1-derived macrophages model. At 24 h after induction of MCAo, hUC-MSCs was administered via tail vein as a single dose. Remarkably, hUC-MSCs could inhibit M1 polarization and promote M2 polarization of microglia in vivo after 14 days induction of MCAo. Compared with THP-1-derived macrophages which had been stimulated by lipopolysaccharide, the secretion of proinflammatory cytokines, tumor necrosis factor-α (TNF-α) and interferon-γ inducible protein (IP-10), were significantly reduced in the presence of hUC-MSCs. Moreover, the secretion of anti-inflammatory cytokine, interleukin-10 (IL-10), was significantly increased after cocultured with hUC-MSCs. Prostaglandins E2 (PGE2), secreted by hUC-MSCs, is one of the crucial immunomodulatory factors and could be inhibited in the presence of COX2 inhibitor, NS-398. PGE2 inhibition suppressed hUC-MSCs immunomodulatory capability, which was restored after addition of synthetic PGE2, establishing the minimum amount of PGE2 required for immunomodulation. In conclusion, our data suggested that PGE2 is a crucial potency marker involved in the therapeutic activity of hUC-MSCs through macrophages immune response modulation and cytokines regulation. This study provides the model for the development of a surrogate quantitative potency assay of immunomodulation in stem cells production.


Assuntos
Isquemia Encefálica/terapia , Dinoprostona/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Isquemia Encefálica/metabolismo , Diferenciação Celular/imunologia , Técnicas de Cocultura/métodos , Citocinas/metabolismo , Dinoprostona/imunologia , Feminino , Sangue Fetal/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Imunidade/efeitos dos fármacos , Imunomodulação/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Microglia/metabolismo , Prostaglandinas E/imunologia , Prostaglandinas E/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Cordão Umbilical/citologia
11.
Int J Mol Med ; 47(1): 207-218, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33416114

RESUMO

Dendritic cells (DCs) are the most potent antigen­presenting cells, and are indispensable in the immune system. Prostaglandin E2 (PGE2) has been demonstrated to modulate the migration of DCs, but with inconsistent results. The present study, based on our previous research, used murine bone marrow­derived DCs to elucidate the potential regulatory mechanism of PGE2 on the migration of DCs. The results indicated that PGE2 served a dual role in regulating the migration of DCs in a dose­dependent manner. High concentrations of PGE2 inhibited cell migration, whereas low concentrations exhibited the opposite effect. Flow cytometry revealed that the expression of CC chemokine receptor type 7 on the DC surface was increased following treatment with low concentrations of PGE2 and slightly decreased by high concentrations of PGE2. The effect of PGE2 was indicated to be exerted via reorganizing the F­actin cytoskeleton using confocal microscopy. Moreover, the regulatory effect of PGE2 on the migration of DCs was validated in vivo. Subsequent gene expression profile analyses using RNA­sequencing technology indicated that PGE2 induced alterations in the expression of multiple downstream genes and signaling pathway molecules associated with cell migration and the cytoskeleton. These findings may provide an improved understanding on the mechanism of DC migration under both pathological and physiological conditions. Moreover, the biological implications of these findings may provide a novel perspective of the immunological surveillance in the progression of different types of diseases.


Assuntos
Movimento Celular/imunologia , Células Dendríticas/imunologia , Dinoprostona/imunologia , Animais , Células Dendríticas/citologia , Masculino , Camundongos
12.
Front Immunol ; 12: 740613, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35126344

RESUMO

The oral mucosa is constantly exposed to a plethora of stimuli including food antigens, commensal microbiota and pathogens, requiring distinct immune responses. We previously reported that human oral epithelial cells (OECs) suppress immune responses to bacteria, using H413 and TR146 OEC lines and primary OECs in co-culture with dendritic cells (DCs) and T cells (OEC-conditioned cells). OECs reduced DCs expression of CD80/CD86 and IL-12/TNFα release and impaired T cell activation. Here, we further evaluated the immunosuppression by these OECs and investigated the underlying mechanisms. OEC-conditioned DCs did not induce CD4 T cell polarization towards Treg, judging by the absence of FoxP3 expression. OECs also repressed T-bet/IFNγ expression in CD4 and CD8 T cells activated by DCs or anti-CD3/CD28 antibodies. This inhibition depended on OEC:T cell ratio and IFNγ repression occurred at the transcriptional level. Time-lapse experiments showed that OECs inhibited early steps of T cell activation, consistent with OECs inability to suppress T cells stimulated with PMA/ionomycin. Blocking CD40/CD40L, CD58/CD2 and PD-L1/PD-1 interactions with specific antibodies did not disrupt T cell suppression by OECs. However, preventing prostaglandin E2 (PGE2) synthesis or blocking PGE2 binding to the cognate EP2/EP4 receptors, restored IFNγ and TNFα production in OEC-conditioned T cells. Finally, treating OECs with poly(I:C), which simulates viral infections, limited T cell suppression. Overall, these results point to an inherent ability of OECs to suppress immune responses, which can nonetheless be eluded when OECs are under direct assault.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Linfócitos T Reguladores/metabolismo , Antígenos CD2/imunologia , Antígenos CD2/metabolismo , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Antígenos CD58/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Dinoprostona/imunologia , Humanos , Tolerância Imunológica/imunologia , Imunidade/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/imunologia , Transcrição Gênica/imunologia
13.
Front Immunol ; 11: 588500, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329572

RESUMO

The results of recent studies have shown that granulocytic-myeloid derived suppressor cells (G-MDSCs) can secrete exosomes that transport various biologically active molecules with regulatory effects on immune cells. However, their roles in autoimmune diseases such as rheumatoid arthritis remain to be further elucidated. In the present study, we investigated the influence of exosomes from G-MDSCs on the humoral immune response in murine collagen-induced arthritis (CIA). G-MDSCs exosomes-treated mice showed lower arthritis index values and decreased inflammatory cell infiltration. Treatment with G-MDSCs exosomes promoted splenic B cells to secrete IL-10 both in vivo and in vitro. In addition, a decrease in the proportion of plasma cells and follicular helper T cells was observed in drainage lymph nodes from G-MDSCs exosomes-treated mice. Moreover, lower serum levels of IgG were detected in G-MDSCs exosomes-treated mice, indicating an alteration of the humoral environment. Mechanistic studies showed that exosomal prostaglandin E2 (PGE2) produced by G-MDSCs upregulated the phosphorylation levels of GSK-3ß and CREB, which play a key role in the production of IL-10+ B cells. Taken together, our findings demonstrated that G-MDSC exosomal PGE2 attenuates CIA in mice by promoting the generation of IL-10+ Breg cells.


Assuntos
Artrite Experimental/imunologia , Linfócitos B/imunologia , Dinoprostona/imunologia , Exossomos/imunologia , Células Supressoras Mieloides/imunologia , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/imunologia , Glicogênio Sintase Quinase 3 beta/imunologia , Granulócitos/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA
14.
Mol Biol Rep ; 47(12): 10015-10021, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33174083

RESUMO

Infection of the uterus with Gram-positive Trueperella pyogenes and Gram-negative Escherichia coli is a common cause of postpartum endometritis in the cattle and buffalo and the condition is treated with antimicrobial drugs. The presence of drug residues in the milk and development of resistant bacteria necessitate the evaluation of alternate therapies for endometritis. Accordingly, we tested the immunomodulatory effect of curcumin in the bubaline endometrial stromal cells after treatment with the lipoteichoic acid (LTA) of Gram-positive Staphylococcus aureus and lipopolysaccharide (LPS) of Gram-negative E. coli that activate toll-like receptors (TLR-2 and TLR-4, respectively). Confluent primary culture of endometrial stromal cells was treated with LTA (1 µg/mL) and/or LPS (0.1 µg/mL), in the presence or absence of curcumin (30 µM for 24 h). PGE2 was assayed in the supernatant and the relative expression of proinflammatory cytokines (PICs) (IL1B, IL6, IL8 and TNFA) transcripts were quantified using real-time PCR. LTA was not effective in stimulating PGE2 production or upregulating the PIC expression except IL8. LTA+LPS increased PGE2 production and upregulated IL6 and IL8 genes. Curcumin inhibited the basal and LTA+LPS induced production of PGE2 and upregulation of PIC production. It was apparent that LPS, but not LTA, is a potent stimulator of PGE2 from the bubaline endometrial stromal cells. Curcumin downregulated the expression of LPS and/or LTA induced PICs and PGE2 and may be an alternate to antimicrobial drugs for the therapeutic management of endometritis.


Assuntos
Búfalos/imunologia , Curcumina , Dinoprostona/imunologia , Endometrite , Endométrio , Células Estromais , Animais , Bovinos , Células Cultivadas , Curcumina/farmacologia , Citocinas/imunologia , Endometrite/tratamento farmacológico , Endometrite/imunologia , Endometrite/veterinária , Endométrio/efeitos dos fármacos , Endométrio/imunologia , Endométrio/patologia , Feminino , Cultura Primária de Células , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Células Estromais/patologia
15.
Bull Exp Biol Med ; 170(1): 49-52, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33222085

RESUMO

We studied the effects of IL-1ß, IL-8, TNFα, and prostaglandin E2α in concentrations typically observed in health and during inflammation on the growth of vaginal microbiota and its resistance to factors inhibiting the synthesis of proteins, nucleic acids, and peptidoglycans. An increase in the cytokine levels, characteristic of inflammation, inhibits the growth of Lactobacillus population and improves its resistance to adverse factors. The growth of the population of opportunistic microorganisms (S. aureus, E. coli) is stimulated under these conditions, while their resistance to adverse factors decreases. Hence, it seems that the cytokines regulate the behavior of the host cells and of its bacterial symbionts.


Assuntos
Dinoprostona/farmacologia , Mediadores da Inflamação/farmacologia , Interleucina-1beta/farmacologia , Interleucina-8/farmacologia , Microbiota/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Vaginose Bacteriana/microbiologia , Líquidos Corporais/microbiologia , Estudos de Casos e Controles , Dinoprostona/imunologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Feminino , Interações Hospedeiro-Patógeno , Humanos , Inflamação , Mediadores da Inflamação/imunologia , Interleucina-1beta/imunologia , Interleucina-8/imunologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/crescimento & desenvolvimento , Microbiota/imunologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Fator de Necrose Tumoral alfa/imunologia , Vagina/imunologia , Vagina/microbiologia , Vaginose Bacteriana/imunologia
16.
Pharmacol Rev ; 72(4): 910-968, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32962984

RESUMO

Prostaglandins are derived from arachidonic acid metabolism through cyclooxygenase activities. Among prostaglandins (PGs), prostacyclin (PGI2) and PGE2 are strongly involved in the regulation of homeostasis and main physiologic functions. In addition, the synthesis of these two prostaglandins is significantly increased during inflammation. PGI2 and PGE2 exert their biologic actions by binding to their respective receptors, namely prostacyclin receptor (IP) and prostaglandin E2 receptor (EP) 1-4, which belong to the family of G-protein-coupled receptors. IP and EP1-4 receptors are widely distributed in the body and thus play various physiologic and pathophysiologic roles. In this review, we discuss the recent advances in studies using pharmacological approaches, genetically modified animals, and genome-wide association studies regarding the roles of IP and EP1-4 receptors in the immune, cardiovascular, nervous, gastrointestinal, respiratory, genitourinary, and musculoskeletal systems. In particular, we highlight similarities and differences between human and rodents in terms of the specific roles of IP and EP1-4 receptors and their downstream signaling pathways, functions, and activities for each biologic system. We also highlight the potential novel therapeutic benefit of targeting IP and EP1-4 receptors in several diseases based on the scientific advances, animal models, and human studies. SIGNIFICANCE STATEMENT: In this review, we present an update of the pathophysiologic role of the prostacyclin receptor, prostaglandin E2 receptor (EP) 1, EP2, EP3, and EP4 receptors when activated by the two main prostaglandins, namely prostacyclin and prostaglandin E2, produced during inflammatory conditions in human and rodents. In addition, this comparison of the published results in each tissue and/or pathology should facilitate the choice of the most appropriate model for the future studies.


Assuntos
Receptores de Prostaglandina E/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Dinoprostona/imunologia , Dinoprostona/metabolismo , Epoprostenol/imunologia , Epoprostenol/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Polimorfismo de Nucleotídeo Único , Multimerização Proteica , Ratos , Receptores de Prostaglandina E/química , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/imunologia , Especificidade da Espécie
17.
Clin Immunol ; 220: 108596, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32961332

RESUMO

Intestinal ischemia/reperfusion (I/R)-induced injury is an inflammatory response with significant morbidity and mortality. The early inflammatory response includes neutrophil infiltration. However, the majority of rodent studies utilize male mice despite a sexual dimorphism in intestinal I/R-related diseases. We hypothesized that sex may alter inflammation by changing neutrophil infiltration and eicosanoid production. To test this hypothesis, male and female C57Bl/6 mice were subjected to sham treatment or 30 min intestinal ischemia followed by a time course of reperfusion. We demonstrate that compared to male mice, females sustain significantly less intestinal I/R-induced tissue damage and produced significant LTB4 concentrations. Male mice release PGE2. Finally, treatment with a COX-2 specific inhibitor, NS-398, attenuated I/R-induced injury, total peroxidase level, and PGE2 production in males, but not in similarly treated female mice. Thus, I/R-induced eicosanoid production and neutrophil infiltration varies between sexes suggesting that distinct therapeutic intervention may be needed in clinical ischemic diseases.


Assuntos
Dinoprostona/imunologia , Leucotrieno B4/imunologia , Mesentério/irrigação sanguínea , Traumatismo por Reperfusão/imunologia , Caracteres Sexuais , Animais , Complemento C5a/imunologia , Citocinas/imunologia , Eicosanoides/imunologia , Feminino , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Peroxidase/imunologia , Traumatismo por Reperfusão/patologia
18.
Vet Immunol Immunopathol ; 228: 110100, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32871408

RESUMO

Mesenchymal stem cells (MSC) are modern tools in regenerative therapies of humans and animals owed to their immunomodulatory properties, which are activated in a pro-inflammatory environment. Different preconditioning strategies had been devised to enhance the immunomodulatory properties of MSC. In this research, we evaluated the immunological attributes of equine adipose MSC (eAMSC) before and after preconditioning in vitro with prostaglandin E2 (PGE2), substance P (SP), their combination and IFNγ. PGE2/SP was the best combination to keep or enhance the mesodermal lineage differentiation of eAMSC. Alongside with this, preconditioning of eMSC with PGE2 and SP did not affect expression of stemness MSC surface phenotype: CD90+, CD44+, MHC class I+, MHC class II- and CD45-, assessed by cytometry. Both naïve and preconditioned eAMSC expressed genes related with immune properties, such as MHC-I, PTGES, IL6, IL1A, TNFα and IL8 assessed by qPCR. Only TNFα was under expressed in treated cells, while the other markers were either overexpressed or not changed. In no cases MHC-II expression was detected. The antiproliferative effect of preconditioned eAMSC exposed to activated peripheral blood mononuclear cells (PBMC) showed that SP treatment significantly inhibited proliferation of LPS stimulated PBMC. When eAMSC were stimulated with Poly I:C, all the treatments significantly inhibited proliferation of stimulated PBMC (p < 0.05). Direct contact (coculture) between the preconditioned eAMSC and PBMC, induced a shift of significantly more (CD4/CD25/FOXP3)+ T-regulatory PBMC than naïve eAMSC. In the experiments of this research, we investigated the secreted proteomic profile of naïve and preconditioned eAMSC, 42 up-regulated and 40 down-regulated proteins were found in the proteomic assay. Our proteomic data revealed profound changes in the secretory pattern of MSC exposed to different treatments, compared to naïve eAMSC as well as among treatments. In overall, compared to naïve cells, the protein profile of preconditioned cells resembled the mesenchymal-epithelial transition (MET). Here we showed that the combined use of PGE2 and SP provoked in overall the highest expression of anti-inflammatory markers as well as lead to an increased acquisition of a T-regulatory phenotype in preconditioned eAMSC without affecting their "stemness".


Assuntos
Dinoprostona/imunologia , Cavalos/imunologia , Células-Tronco Mesenquimais/imunologia , Proteínas/metabolismo , Substância P/imunologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular/imunologia , Proliferação de Células , Citometria de Fluxo/veterinária , Interferon gama/imunologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Mesoderma/citologia , Proteoma , Via Secretória/imunologia
19.
J Food Sci ; 85(9): 2822-2831, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32794226

RESUMO

The anti-inflammatory effects of two newly identified Pleurotus eryngii polysaccharides (WPEP, NPEP) were determined in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages in this study. Characterization analysis revealed that molecular weights of WPEP and NPEP were 167 and 274 kDa, and were mainly composed of glucose with ß-type glycosidic linkages. WPEP and NPEP could significantly inhibit LPS-induced inflammatory responses by regulating the production of NO, Protaglandin E2 (PGE2 ), Interleukin-1ß (IL-1ß), Tumor necrosis factor-α (TNF-α), and Interleukin-6 (IL-6). This was through the blocking of the activation of Mitogen-activated protein kinase (MAPK) pathway by inhibiting phosphorylation of p38, extracellular regulation of protein kinases 1/2, and stress-activated protein kinase/jun aminoterminal kinase. Moreover, WPEP and NPEP inhibited NF-κB signaling by reducing nuclear translocation and phosphorylation of p65. Overall, our results, for the first time identified two P. eryngii polysaccharides and demonstrated the related anti-inflammatory effects, which indicated the favorable potential of P. eryngii polysaccharide as specific functional foods. PRACTICAL APPLICATION: This study prepared and characterized newly identified Pleurotus eryngii water-soluble polysaccharide fractions and elucidated the nutritional benefits, mainly the immune response related to anti-inflammatory activities by utilizing lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Collectively, results of this study suggested that the P. eryngii polysaccharide fractions could be considered as potential candidates for exploration in the development of new immunomodulatory agent or functional supplementary foods.


Assuntos
Anti-Inflamatórios/farmacologia , Extratos Vegetais/farmacologia , Pleurotus/química , Polissacarídeos/farmacologia , Animais , Anti-Inflamatórios/química , Dinoprostona/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , Extratos Vegetais/química , Polissacarídeos/química , Células RAW 264.7 , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
20.
Front Immunol ; 11: 1224, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32636842

RESUMO

Increased serum IgE level is one of the features of allergic asthma. It is reported that IgE production can be enhanced by E-prostanoid 2 (EP2) receptor of prostaglandin E2 (PGE2); however, whether E-prostanoid 4 (EP4) receptor (encoded by Ptger4) has a unique or redundant role is still unclear. Here, we demonstrated the mice with B cell-specific deletion of the EP4 receptor (Ptger4fl/flMb1cre+/-) showed their serum levels of IgE were markedly increased. A much more severe airway allergic inflammation was observed in the absence of EP4 signal using the OVA-induced asthma model. Mechanistic studies demonstrated that the transcription levels of AID, GLTε, and PSTε in EP4-deficient B cells were found to be significantly increased, implying an enhanced IgE class switch. In addition, we saw higher levels of phosphorylated STAT6, a vital factor for IgE class switch. Biochemical analyses indicated that inhibitory effect of EP4 signal on IgE depended on the activation of the PI3K-AKT pathway. Further downstream, PPARγ expression was up-regulated. Independent of its activity as a transcription factor, PPARγ here primarily functioned as an E3 ubiquitin-ligase, which bound the phosphorylated STAT6 to initiate its degradation. In support of PPARγ as a key mediator downstream of the EP4 signal, PPARγ agonist induced the down-regulation of phospho-STAT6, whereas its antagonist was able to rescue the EP4-mediated inhibition of STAT6 activation and IgE production. Thus, our findings highlight a role for the PGE2-EP4-AKT-PPARγ-STAT6 signaling in IgE response, highlighting the therapeutic potential of combined application of EP4 and PPARγ agonists in asthma.


Assuntos
Asma/metabolismo , Dinoprostona/metabolismo , Switching de Imunoglobulina/imunologia , Imunoglobulina E/biossíntese , PPAR gama/metabolismo , Animais , Asma/imunologia , Dinoprostona/imunologia , Imunoglobulina E/imunologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/imunologia , Estabilidade Proteica , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/imunologia , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...