Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
1.
Nat Commun ; 12(1): 6798, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34815397

RESUMO

G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Receptores Purinérgicos P2Y/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/metabolismo , Adulto , Idoso , Animais , Comunicação Autócrina/imunologia , Linfócitos T CD4-Positivos/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Quimiocinas/metabolismo , Quimiotaxia de Leucócito/imunologia , Encefalomielite Autoimune Experimental/sangue , Feminino , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Humanos , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Esclerose Múltipla/sangue , Comunicação Parácrina/imunologia , Cultura Primária de Células , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y/genética , Proteína rhoA de Ligação ao GTP/metabolismo
3.
FEBS J ; 288(2): 405-426, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32502309

RESUMO

The interface between the mammalian host and its environment is formed by barrier tissues, for example, of the skin, and the respiratory and the intestinal tracts. On the one hand, barrier tissues are colonized by site-adapted microbial communities, and on the other hand, they contain specific myeloid cell networks comprising macrophages, dendritic cells, and granulocytes. These immune cells are tightly regulated in function and cell number, indicating important roles in maintaining tissue homeostasis and immune balance in the presence of commensal microorganisms. The regulation of myeloid cell density and activation involves cell-autonomous 'single-loop circuits' including autocrine mechanisms. However, an array of microenvironmental factors originating from nonimmune cells and the microbiota, as well as the microanatomical structure, impose additional layers of regulation onto resident myeloid cells. This review discusses models integrating these factors into cell-specific programs to instruct differentiation and proliferation best suited for the maintenance and renewal of immune homeostasis in the tissue-specific environment.


Assuntos
Células Dendríticas/imunologia , Granulócitos/microbiologia , Macrófagos/imunologia , Microbiota/fisiologia , Modelos Imunológicos , Simbiose/imunologia , Animais , Comunicação Autócrina/imunologia , Contagem de Células , Diferenciação Celular , Proliferação de Células , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/microbiologia , Granulócitos/imunologia , Homeostase/imunologia , Humanos , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Macrófagos/microbiologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Pele/imunologia , Pele/microbiologia
4.
Cell Mol Gastroenterol Hepatol ; 11(1): 249-272, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32827707

RESUMO

BACKGROUND & AIMS: TNFSF15 genetic variants leading to increased TNF superfamily member 15 (TNFSF15) expression confer risk for inflammatory bowel disease (IBD), and TNFSF15 is being explored as a therapeutic target in IBD patients. Although the focus for TNFSF15-mediated inflammatory outcomes has been predominantly on its action on T cells, TNFSF15 also promotes inflammatory outcomes in human macrophages. Given the critical role for macrophages in bacterial clearance, we hypothesized that TNFSF15 promotes antimicrobial pathways in human macrophages and that macrophages from TNFSF15 IBD risk carriers with higher TNFSF15 expression have an advantage in these antimicrobial outcomes. METHODS: We analyzed protein expression, signaling, bacterial uptake, and intracellular bacterial clearance in human monocyte-derived macrophages through flow cytometry, enzyme-linked immunosorbent assay, and gentamicin protection. RESULTS: Autocrine/paracrine TNFSF15 interactions with death receptor 3 (DR3) were required for optimal levels of pattern-recognition-receptor (PRR)-induced bacterial clearance in human macrophages. TNFSF15 induced pyruvate dehydrogenase kinase 1-dependent bacterial uptake and promoted intracellular bacterial clearance through reactive oxygen species, nitric oxide synthase 2, and autophagy up-regulation. The TNFSF15-initiated TNF receptor-associated factor 2/receptor-interacting protein kinase 1/RIP3 pathway was required for mitogen-activated protein kinase and nuclear factor-κB activation, and, in turn, induction of each of the antimicrobial pathways; the TNFSF15-initiated Fas-associated protein with death domain/mucosa-associated lymphoid tissue lymphoma translocation protein 1/caspase-8 pathway played a less prominent role in antimicrobial functions, despite its key role in TNFSF15-induced cytokine secretion. Complementation of signaling pathways or antimicrobial pathways restored bacterial uptake and clearance in PRR-stimulated macrophages where TNFSF15:DR3 interactions were inhibited. Monocyte-derived macrophages from high TNFSF15-expressing rs6478108 TT IBD risk carriers in the TNFSF15 region showed increased levels of the identified antimicrobial pathways. CONCLUSIONS: We identify that autocrine/paracrine TNFSF15 is required for optimal PRR-enhanced antimicrobial pathways in macrophages, define mechanisms regulating TNFSF15-dependent bacterial clearance, and determine how the TNFSF15 IBD risk genotype modulates these outcomes.


Assuntos
Doenças Inflamatórias Intestinais/imunologia , Macrófagos/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Comunicação Autócrina/imunologia , Células Cultivadas , Enterococcus faecalis/imunologia , Enterococcus faecalis/isolamento & purificação , Escherichia coli/imunologia , Escherichia coli/isolamento & purificação , Predisposição Genética para Doença , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/microbiologia , Macrófagos/metabolismo , Camundongos , Comunicação Parácrina/imunologia , Cultura Primária de Células , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
5.
J Invest Dermatol ; 141(3): 596-606.e7, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32853659

RESUMO

IL-33 is constitutively expressed in the skin. Psoriasis is a common skin inflammatory disease. The roles of IL-33 in psoriasis have not been well-elucidated. We identified that keratinocytes (KCs) are the predominant cells expressing IL-33 and its receptor, suppression of tumorigenicity 2, in the skin. KCs actively released IL-33 on psoriasis inflammatory stimuli and induced psoriasis-related cytokine, chemokine, and inflammatory molecules genes transcription in KCs in an autocrine manner. IL-33‒specific deficiency in KCs ameliorated imiquimod-induced psoriatic dermatitis. In addition, intradermal injection of recombinant IL-33 alone induced psoriasis-like dermatitis, which is attributed to the transcriptional upregulation of genes enriched in IL-17, TNF, and chemokine signaling pathway in KCs on recombinant IL-33 stimulation. Our data demonstrate that the autocrine circuit of IL-33 in KCs promotes the progression of psoriatic skin inflammation, and IL-33 is a potential therapeutic target for psoriasis.


Assuntos
Interleucina-33/metabolismo , Queratinócitos/metabolismo , Psoríase/imunologia , Adulto , Animais , Comunicação Autócrina/imunologia , Biópsia , Estudos de Casos e Controles , Modelos Animais de Doenças , Progressão da Doença , Voluntários Saudáveis , Humanos , Imiquimode/imunologia , Injeções Intradérmicas , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/administração & dosagem , Interleucina-33/genética , Queratinócitos/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Psoríase/diagnóstico , Psoríase/genética , Psoríase/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Pele/imunologia , Pele/patologia , Ativação Transcricional/imunologia , Regulação para Cima/imunologia
6.
Int J Mol Sci ; 21(18)2020 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-32971928

RESUMO

Semaphorin (Sema)4A is a transmembrane glycoprotein that is elevated in several autoimmune diseases such as systemic sclerosis, rheumatoid arthritis and multiple sclerosis. Sema4A has a key role in the regulation of Thelper Th1 and Th2 differentiation and we recently demonstrated that CD4+ T cell activation induces the expression of Sema4A. However, the autocrine role of Sema4A on Th cell differentiation remains unknown. Naïve Th cells from healthy controls were cell sorted and differentiated into Th1, Th2 and Th17 in the presence or absence of a neutralizing antibody against the Sema4A receptor PlexinD1. Gene expression was determined by quantitative PCR and protein expression by ELISA and flow cytometry. We found that the expression of Sema4A is induced during Th1, Th2 and Th17 differentiation. PlexinD1 neutralization induced the differentiation of Th1 cells, while reduced the Th2 and Th17 skewing. These effects were associated with an upregulation of the transcription factor T-bet by Th1 cells, and to downregulation of GATA3 and RORγt in Th2 cells and Th17 cells, respectively. Finally, PlexinD1 neutralization regulates the systemic sclerosis patients serum-induced cytokine production by CD4+ T cells. Therefore, the autocrine Sema4A-PlexinD1 signaling acts as a negative regulator of Th1 skewing but is a key mediator on Th2 and Th17 differentiation, suggesting that dysregulation of this axis might be implicated in the pathogenesis of CD4+ T cell-mediated diseases.


Assuntos
Comunicação Autócrina/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Glicoproteínas de Membrana/imunologia , Semaforinas/imunologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/patologia , Células Th1/patologia , Células Th17/patologia , Células Th2/patologia
7.
J Autoimmun ; 115: 102540, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32893081

RESUMO

Lupus nephritis (LN) is a major cause for overall morbidity and mortality in patients with systemic lupus erythematosus (SLE), while its pathogenic mechanisms are still not well understood. Extracellular vesicles (EVs) are membrane vesicles that are released from almost all cell types. EVs can be subdivided into exosomes, microvesicles, and apoptotic bodies. Latest studies have shown that EVs can be released during several cellular events, including cell activation, autophagy, and several types of programed cell death, i.e. apoptosis, necroptosis, pyroptosis, and NETosis. Emerging evidence demonstrates that EVs harbor different bioactive molecules, including nucleic acids, proteins, lipids, cytokines, immune complexes (ICs), complements, and other molecules, some of which may contribute to pathogenesis of autoimmune diseases. EVs can serve as novel information shuttle to mediate local autocrine or paracrine signals to nearby cells, and distant endocrine signals to cells located far away. In LN, EVs may have pathogenic effects by transportation of autoantigens or complements, promotion of IC deposition or complement activation, and stimulation of inflammatory responses, renal tissue injury, or microthrombus formation. Additionally, EVs released from kidney cells may serve as specific biomarkers for diagnosis or monitoring of disease activity and therapeutic efficacy. In this review, we will summarize the latest progress about EV generation from basic research, their potential pathologic effects on LN, and their clinical implications. The cutting-edge knowledge about EV research provides insights into novel therapeutic strategy, new tools for diagnosis or prognosis, and evaluation approaches for treatment effectiveness in LN.


Assuntos
Vesículas Extracelulares/imunologia , Rim/patologia , Nefrite Lúpica/imunologia , Comunicação Autócrina/imunologia , Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Rim/citologia , Rim/imunologia , Nefrite Lúpica/sangue , Nefrite Lúpica/diagnóstico , Nefrite Lúpica/patologia , Comunicação Parácrina/imunologia , Prognóstico , Índice de Gravidade de Doença
8.
Oncogene ; 39(40): 6354-6369, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32855524

RESUMO

In patients with lung cancer, myeloid-derived suppressor cells (MDSCs) have been reported to be significantly increased. Tumor-derived exosomes (TDEs) from various cancers played a critical role in MDSC induction. However, studies on the molecular mechanism underlying MDSC expansion induced by exosomes from lung cancer cells are still limited. In this study, we demonstrated that LLC-Exo accelerated tumor growth along with a significant accumulation of MDSCs in mouse tumor model. miRNA profiling showed that miR-21a was enriched in LLC-Exo. The depletion of miR-21a in LLC-Exo leads to the loss of their ability to induce MDSC expansion. Further results showed that miR-21a of LLC-Exo induced MDSC expansion via downregulation of the programmed cell death protein 4 (PDCD4) protein. The results of gain-and loss-of-function experiments validated that PDCD4 function as a critical inhibitor to negatively regulate expansion of MDSCs via inhibition Il-6 production in bone marrow cells. In addition, our data showed that exosomes derived from human lung cancer cell lines expressing miR-21a, also induced expansion of MDSCs in human CD14+ monocytes in vitro. Overall, our results demonstrated that miR-21a enriched in lung carcinoma cell-derived exosomes could promote functional expansion of MDSCs through targeting PDCD4.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Carcinoma Pulmonar de Lewis/imunologia , MicroRNAs/metabolismo , Células Supressoras Mieloides/imunologia , Proteínas de Ligação a RNA/genética , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Comunicação Autócrina/genética , Comunicação Autócrina/imunologia , Carcinoma Pulmonar de Lewis/genética , Linhagem Celular Tumoral/transplante , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Regulação para Baixo , Exossomos/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Voluntários Saudáveis , Humanos , Interleucina-6/metabolismo , Leucócitos Mononucleares , Camundongos , MicroRNAs/agonistas , MicroRNAs/genética , Células Supressoras Mieloides/metabolismo , Fosforilação/genética , Fosforilação/imunologia , Cultura Primária de Células , Proteínas de Ligação a RNA/metabolismo , Fator de Transcrição STAT3/metabolismo
9.
Integr Biol (Camb) ; 12(7): 175-187, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32556172

RESUMO

Hematopoietic stem cells (HSCs) primarily reside in the bone marrow, where they receive external cues from their local microenvironment. The complex milieu of biophysical cues, cellular components and cell-secreted factors regulates the process by which HSC produce the blood and immune system. We previously showed direct coculture of primary murine hematopoietic stem and progenitor cells with a population of marrow-derived mesenchymal stromal and progenitor cells (MSPCs) in a methacrylamide-functionalized gelatin (GelMA) hydrogel improves hematopoietic progenitor maintenance. However, the mechanism by which MSPCs influenced HSC fate decisions remained unknown. Herein, we report the use of proteomic analysis to correlate HSC phenotype to a broad candidate pool of 200 soluble factors produced by combined mesenchymal and hematopoietic progeny. Partial least squares regression (PLSR), along with an iterative filter method, identified TGFß-1, MMP-3, c-RP and TROY as positively correlated with HSC maintenance. Experimentally, we then observe exogenous stimulation of HSC monocultures in GelMA hydrogels with these combined cytokines increases the ratio of hematopoietic progenitors to committed progeny after a 7-day culture 7.52 ± 3.65-fold compared to non-stimulated monocultures. Findings suggest a cocktail of the downselected cytokines amplifies hematopoietic maintenance potential of HSCs beyond that of MSPC-secreted factors alone. This work integrates empirical and computation methods to identify cytokine combinations to improve HSC maintenance within an engineered HSC niche, suggesting a route toward identifying feeder-free culture platforms for HSC expansion. Insight Hematopoietic stem cells within an artificial niche receive maintenance cues in the form of soluble factors from hematopoietic and mesenchymal progeny. Applying a proteomic regression analysis, we identify a reduced set of soluble factors correlated to maintenance of a hematopoietic phenotype during culture in a biomaterial model of the bone marrow niche. We identify a minimum factor cocktail that promotes hematopoietic maintenance potential in a gelatin-based culture, regardless of the presence of mesenchymal feeder cells. By combining empirical and computational methods, we report an experimentally feasible number of factors from a large dataset, enabling exogenous integration of soluble factors into an engineered hematopoietic stem cell for enhanced maintenance potential of a quiescent stem cell population.


Assuntos
Medula Óssea/metabolismo , Engenharia Celular , Células-Tronco Hematopoéticas/metabolismo , Nicho de Células-Tronco/genética , Algoritmos , Animais , Comunicação Autócrina/imunologia , Células Cultivadas , Citocinas/metabolismo , Feminino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Comunicação Parácrina/imunologia , Fenótipo , Proteômica/métodos
10.
Front Immunol ; 11: 540, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32300344

RESUMO

The ability of T cells to sense and respond to environmental cues by altering their functional capabilities is critical for a safe and optimally protective immune response. One of the important properties that contributes to this goal is the activation set-point of the T cell. Here we report a new pathway through which TCR transgenic OT-I CD8+ T cells can self-tune their activation threshold. We find that in the presence of a strong TCR engagement event there is a shift in the metabolic programming of the cell where both glycolysis and oxidative phosphorylation are significantly increased. This diverges from the switch to a predominantly glycolytic profile that would be predicted following naïve T cell activation. Our data suggest this altered metabolic program results in the production of autocrine IL-4. Both metabolic pathways are required for this cytokine to be made. IL-4 signaling in the activated OT-I CD8+ T cell results in modulation of the sensitivity of the cell, establishing a higher activation setpoint that is maintained over time. Together these data demonstrate a novel mechanism for the regulation of IL-4 production in CD8+ T cells. Further, they reveal a new pathway for the self-tuning of peptide sensitivity. Finally, these studies uncover an unexpected role for oxidative phosphorylation in regulating differentiation in these cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Interleucina-4/biossíntese , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Comunicação Autócrina/imunologia , Camundongos , Camundongos Endogâmicos C57BL
11.
Gut ; 69(2): 264-273, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31097538

RESUMO

OBJECTIVE: The interleukin (IL)23 pathway contributes to IBD pathogenesis and is being actively studied as a therapeutic target in patients with IBD. Unexpected outcomes in these therapeutic trials have highlighted the importance of understanding the cell types and mechanisms through which IL23 regulates immune outcomes. How IL23 regulates macrophage outcomes and the consequences of the IL23R R381Q IBD-protective variant on macrophages are not well defined; macrophages are key players in IBD pathogenesis and inflammation. DESIGN: We analysed protein and RNA expression, signalling and localisation in human monocyte-derived macrophages (MDMs) through western blot, ELISA, real-time PCR, flow cytometry, immunoprecipitation and microscopy. RESULTS: IL23R was critical for optimal levels of pattern-recognition receptor (PRR)-induced signalling and cytokines in human MDMs. In contrast to the coreceptor IL12Rß1, IL23 induced dynamic IL23R cell surface regulation and this required clathrin and dynamin-mediated endocytosis and endocytic recycling-dependent pathways; these pathways were essential for IL23R-mediated outcomes. The IBD-protective IL23R R381Q variant showed distinct outcomes. Relative to IL23R R381, HeLa cells expressing IL23R Q381 showed decreased IL23R recycling and reduced assembly of IL23R Q381 with Janus kinase/signal transducer and activator of transcription pathway members. In MDMs from IL23R Q381 carriers, IL23R accumulated in late endosomes and lysosomes on IL23 treatment and cells demonstrated decreased IL23R- and PRR-induced signalling and cytokines relative to IL23R R381 MDMs. CONCLUSION: Macrophage-mediated inflammatory pathways are key contributors to IBD pathogenesis, and we identify an autocrine/paracrine IL23 requirement in PRR-initiated human macrophage outcomes and in human intestinal myeloid cells, establish that IL23R undergoes ligand-induced recycling, define mechanisms regulating IL23R-induced signalling and determine how the IBD-protective IL23R R381Q variant modulates these processes.


Assuntos
Citocinas/imunologia , Doenças Inflamatórias Intestinais/imunologia , Macrófagos/imunologia , Receptores de Interleucina/imunologia , Comunicação Autócrina/imunologia , Endocitose/imunologia , Endossomos/imunologia , Variação Genética , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/prevenção & controle , Interleucina-23/imunologia , Janus Quinase 2/metabolismo , Comunicação Parácrina/imunologia , Receptores de Interleucina/genética , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/imunologia
12.
JCI Insight ; 4(18)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31534048

RESUMO

With multifactorial etiologies, combined with disease heterogeneity and a lack of suitable diagnostic markers and therapy, endometriosis remains a major reproductive health challenge. Extracellular vesicles (EVs) have emerged as major contributors of disease progression in several conditions, including a variety of cancers; however, their role in endometriosis pathophysiology has remained elusive. Using next-generation sequencing of EVs obtained from endometriosis patient tissues and plasma samples compared with controls, we have documented that patient EVs carry unique signatures of miRNAs and long noncoding RNAs (lncRNAs) reflecting their contribution to disease pathophysiology. Mass spectrophotometry-based proteomic analysis of EVs from patient plasma and peritoneal fluid further revealed enrichment of specific pathways, as well as altered immune and metabolic processes. Functional studies in endometriotic epithelial and endothelial cell lines using EVs from patient plasma and controls clearly indicate autocrine uptake and paracrine cell proliferative roles, suggestive of their involvement in endometriosis. Multiplex cytokine analysis of cell supernatants in response to patient and control plasma-derived EVs indicate robust signatures of important inflammatory and angiogenic cytokines known to be involved in disease progression. Collectively, these findings suggest that endometriosis-associated EVs carry unique cargo and contribute to disease pathophysiology by influencing inflammation, angiogenesis, and proliferation within the endometriotic lesion microenvironment.


Assuntos
Endometriose/genética , Exossomos/genética , Inflamação/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Líquido Ascítico/citologia , Comunicação Autócrina/genética , Comunicação Autócrina/imunologia , Estudos de Casos e Controles , Linhagem Celular , Proliferação de Células/genética , Citocinas/genética , Citocinas/imunologia , Progressão da Doença , Endometriose/sangue , Endometriose/imunologia , Endometriose/patologia , Endométrio/irrigação sanguínea , Endométrio/citologia , Endométrio/patologia , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Regulação da Expressão Gênica/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/patologia , Microscopia Intravital , MicroRNAs/isolamento & purificação , Microscopia Eletrônica de Transmissão , Neovascularização Patológica/genética , Comunicação Parácrina/genética , Comunicação Parácrina/imunologia , Proteômica , RNA Longo não Codificante/isolamento & purificação , RNA-Seq
13.
J Immunol ; 203(2): 379-388, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31217324

RESUMO

The involvement of complement in B2 cell responses has been regarded as occurring strictly via complement components in plasma. In this study, we show that Ab production and class switch recombination (CSR) depend on autocrine C3a and C5a receptor (C3ar1/C5ar1) signaling in B2 cells. CD40 upregulation, IL-6 production, growth in response to BAFF or APRIL, and AID/Bcl-6 expression, as well as follicular CD4+ cell CD21 production, all depended on this signal transduction. OVA immunization of C3ar1-/-C5ar1-/- mice elicited IgM Ab but no other isotypes, whereas decay accelerating factor (Daf1)-/- mice elicited more robust Ab production and CSR than wild-type (WT) mice. Comparable differences occurred in OVA-immunized µMT recipients of WT, C3ar1-/-C5ar1-/- , and Daf1-/- B2 cells and in hen egg lysozyme-immunized µMT recipients of MD4 B2 cells on each genetic background. B2 cells produced factor I and C3 and autophosphorylated CD19. Immunized C3-/-C5-/- recipients of WT MD4 bone marrow efficiently produced Ab. Thus, B2 cell-produced complement participates in B2 cell activation.


Assuntos
Comunicação Autócrina/imunologia , Receptor da Anafilatoxina C5a/imunologia , Receptores de Complemento/imunologia , Animais , Antígenos CD19/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Proteínas do Sistema Complemento/imunologia , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia
14.
Respir Med ; 150: 136-140, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30961940

RESUMO

BACKGROUND: We hypothesized that the peculiar mixed interleukin-4 (IL-4/Th2) and interferon gamma INF-γ (INF-γ/Th1) inflammatory milieu found in the airways of patients with aspirin-exacerbated respiratory disease (AERD) is responsible for the altered regulation of the IL-1ß/IL-1RI-/EP2/COX-2 autocrine loop also found in these patients. The objective of the study is to demonstrate that IL-4 and INF-γ cytokines, are capable of inducing in healthy nasal mucosa (NM) the dysregulation of the autocrine loop of COX reported in AERD. SUBJECTS AND METHODS: Fibroblasts were obtained from NM (n = 8). To evaluate the role of IL-4 and IFN-γ on the autocrine loop, fibroblasts were incubated with or without IL-1ß, in the presence or absence of IL-4 and/or IFN-γ for 48 h. After this period, the expression of EP2, EP3, EP4, IL-1RI, COX-2 and mPGES-1 was measured by Western blot. RESULTS: Stimulation of fibroblasts with IL-1ß significantly increased the expression of EP2, but had no effects on EP3 and EP4 expression Incubation with IL-4 or IFN-γ alone was not able to modify the expression of any of the components of the autocrine loop. In contrast, co-treatment with IL-4 and IFN-γ was able to significantly inhibit IL-1ß-induced EP2, IL-1RI, COX-2 and mPGES-1. CONCLUSION: These results suggest that the mixed Th1/Th2 inflammatory pattern found in the airways of AERD patients might be responsible for the altered regulation of the COX pathway also reported in these asthma patients.


Assuntos
Asma Induzida por Aspirina/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica/imunologia , Mucosa Nasal/metabolismo , Adulto , Comunicação Autócrina/imunologia , Ciclo-Oxigenase 2 , Feminino , Fibroblastos/metabolismo , Humanos , Interferon gama , Interleucina-1beta , Interleucina-4 , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/citologia , Receptores Tipo I de Interleucina-1 , Células Th1/metabolismo , Células Th2/metabolismo
15.
J Immunol ; 202(9): 2609-2615, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30902899

RESUMO

Cholecystokinin (CCK) is a peptide hormone that functions in digestive organs and the CNS. We previously showed that CCK downregulates peripheral pruritus by suppressing degranulation of mast cells. In this study, we demonstrated that CCK octapeptide (CCK8) was constitutively expressed in the epidermis of normal skin, whereas its expression was lost in acanthotic lesions of psoriasis. In contrast, CCKA receptor (CCKAR), a high-affinity receptor for CCK, was constitutively expressed in the epidermis of psoriatic skin lesions. Expression of CCK was also reduced in skin lesions of an imiquimod (IMQ)-induced psoriatic mouse model. Notably, the expression level of CCK inversely correlated with the severity of epidermal inflammation, raising the possibility that CCK from epidermal keratinocytes suppresses the psoriatic inflammation. To verify this hypothesis, we investigated the effects of sulfated CCK octapeptide (CCK8S) on the development of IMQ-induced psoriatic inflammation. i.p. injection of CCK8S suppressed the IMQ-induced psoriatic inflammation accompanied by reduced mRNA expression of IL-17, IL-22, and IL-6 but not of IL-23. The suppressive effect of CCK8S was completely restored by administration of CCKAR antagonist. In vitro studies showed that exogenous CCK8S suppressed IL-6 production in CCKAR-expressing cultured human keratinocytes, and blocking the endogenous CCK signaling with CCKAR antagonist markedly enhanced IL-6 production. When keratinocytes were stimulated with IL-17, the expression of endogenous CCK was significantly decreased. These findings suggest that CCK physiologically functions as a negative regulator of keratinocyte-based inflammation in an autocrine or paracrine manner, although decreased CCK may pathologically contribute to continuous and aggravated skin lesions such as psoriasis.


Assuntos
Colecistocinina/imunologia , Regulação para Baixo/imunologia , Epiderme/imunologia , Queratinócitos/imunologia , Psoríase/imunologia , Transdução de Sinais/imunologia , Animais , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/imunologia , Epiderme/patologia , Feminino , Humanos , Imiquimode/farmacologia , Inflamação/imunologia , Inflamação/patologia , Interleucina-17/imunologia , Interleucina-6/imunologia , Queratinócitos/patologia , Masculino , Camundongos , Oligopeptídeos/imunologia , Oligopeptídeos/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/imunologia , Psoríase/patologia
16.
J Immunol ; 202(8): 2348-2359, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30833347

RESUMO

The type I IFNs (IFN-α and -ß) are important for host defense against viral infections. In contrast, their role in defense against nonviral pathogens is more ambiguous. In this article, we report that IFN-ß signaling in murine bone marrow-derived macrophages has a cell-intrinsic protective capacity against Mycobacterium tuberculosis via the increased production of NO. The antimycobacterial effects of type I IFNs were mediated by direct signaling through the IFN-α/ß-receptor (IFNAR), as Ab-mediated blocking of IFNAR1 prevented the production of NO. Furthermore, M. tuberculosis is able to inhibit IFNAR-mediated cell signaling and the subsequent transcription of 309 IFN-ß-stimulated genes in a dose-dependent way. The molecular mechanism of inhibition by M. tuberculosis involves reduced phosphorylation of the IFNAR-associated protein kinases JAK1 and TYK2, leading to reduced phosphorylation of the downstream targets STAT1 and STAT2. Transwell experiments demonstrated that the M. tuberculosis-mediated inhibition of type I IFN signaling was restricted to infected cells. Overall, our study supports the novel concept that M. tuberculosis evolved to inhibit autocrine type I IFN signaling to evade host defense mechanisms.


Assuntos
Comunicação Autócrina/imunologia , Interferon Tipo I/imunologia , Viabilidade Microbiana/imunologia , Mycobacterium tuberculosis/imunologia , Transdução de Sinais/imunologia , Animais , Comunicação Autócrina/genética , Interferon Tipo I/genética , Janus Quinase 1/genética , Janus Quinase 1/imunologia , Camundongos , Camundongos Knockout , Viabilidade Microbiana/genética , Óxido Nítrico/genética , Óxido Nítrico/imunologia , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/genética , TYK2 Quinase/genética , TYK2 Quinase/imunologia
17.
Eur J Immunol ; 49(3): 386-397, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30443903

RESUMO

Neutrophils destroy invading microorganisms by phagocytosis by bringing them into contact with bactericidal substances, among which ROS are the most important. However, ROS also function as important physiological regulators of cellular signaling pathways. Here, we addressed the involvement of oxygen derivatives in the regulation of human neutrophil rolling, an essential component of the inflammatory response. Flow experiments using dihydroethidium-preloaded human neutrophils showed that these cells initiate an early production of intracellular ROS during the rolling phase of the adhesion cascade, a phenomenon that required cell rolling, and the interaction of the chemokine receptor CXCR2 with their ligand CXCL8. Flow cytometry experiments demonstrated that L-selectin shedding in neutrophils is triggered by ROS through an autocrine-paracrine mechanism. Preincubation of neutrophils with the NADPH oxidase complex inhibitor diphenyleniodonium chloride significantly increased the number of rolling neutrophils on endothelial cells. Interestingly, the same effect was observed when CXCL8 signaling was interfered using either a blocking monoclonal antibody or an inhibitor of its receptor. These findings indicate that, in response to CXCL8, neutrophils initiate ROS production during the rolling phase of the inflammatory response. This very early ROS production might participate in the modulation of the inflammatory response by inducing L-selectin shedding in neutrophils.


Assuntos
Adesão Celular/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Interleucina-8/imunologia , Selectina L/imunologia , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/imunologia , Comunicação Autócrina/imunologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Interleucina-8/metabolismo , Selectina L/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Oxidantes/farmacologia , Comunicação Parácrina/imunologia , Ligação Proteica/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Interleucina-8B/imunologia , Receptores de Interleucina-8B/metabolismo
18.
J Cell Mol Med ; 23(2): 1257-1267, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30456891

RESUMO

M2-polarized macrophages, on one hand, can promote tumour vascularization by producing proangiogenic factors, such as vascular endothelial growth factor (VEGF). On the other hand, the expression of VEGF receptors (VEGFR) in this cell lineage was also reported. Although the function of VEGF/VEGFR axis plays a pivotal role in macrophages infiltration and angiogenesis, however, there is still lack of the direct evidence to show the role of VEGF as an autocrine operating in M2 macrophages, particularly for immunomodulation. In our study, we surprisingly discovered that M2 macrophages polarized by baicalin can simultaneously express VEGF and its receptors. Taking advantage of this unique culture system, we were able to investigate the biological activity of M2 macrophages in response to the autocrine VEGF milieu. Our results showed that the expression of programmed death-ligand 1 (PD-L1) on M2 macrophages was significantly up-regulated in autocrine VEGF milieu. Through the blockade of autocrine VEGF signalling, PD-L1 expression on M2 macrophages was dramatically down-regulated. Furthermore, transplantation of PD-L1+ M2 macrophage stimulated by autocrine VEGF into allogeneic mice significantly suppressed host CD4+ /CD8+ T cells in the peripheral blood and increased CD4+ CD25+ regulatory T cells in the bone marrow. In conclusion, our findings provide a novel biological basis to support the current successful strategy using combined VEGF/PD-1 signalling blockade in cancer therapy.


Assuntos
Comunicação Autócrina/imunologia , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Macrófagos/imunologia , Linfócitos T Reguladores/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/imunologia , Animais , Axitinibe/farmacologia , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Feminino , Flavonoides/farmacologia , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Células RAW 264.7 , Transdução de Sinais , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Transplante Homólogo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
19.
Front Immunol ; 9: 2818, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555483

RESUMO

ß-Glucan derived from cell walls of Candida albicans is a potent immune modulator. It has been shown to induce trained immunity in monocytes via epigenetic and metabolic reprogramming and to protect from lethal sepsis if applied prior to infection. Since ß-glucan-trained monocytes have not been classified within the system of mononuclear phagocytes we analyzed these cells metabolically, phenotypically and functionally with a focus on monocyte-to-macrophage differentiation and compared them with naïve monocytes and other types of monocyte-derived cells such as classically (M1) or alternatively (M2) activated macrophages and monocyte-derived dendritic cells (moDCs). We show that ß-glucan inhibits spontaneous apoptosis of monocytes independent from autocrine or paracrine M-CSF release and stimulates monocyte differentiation into macrophages. ß-Glucan-differentiated macrophages exhibit increased cell size and granularity and enhanced metabolic activity when compared to naïve monocytes. Although ß-glucan-primed cells expressed markers of alternative activation and secreted higher levels of IL-10 after lipopolysaccharide (LPS), their capability to release pro-inflammatory cytokines and to kill bacteria was unaffected. Our data demonstrate that ß-glucan priming induces a population of immune competent long-lived monocyte-derived macrophages that may be involved in immunoregulatory processes.


Assuntos
Candida albicans/química , Diferenciação Celular/efeitos dos fármacos , Macrófagos/imunologia , Monócitos/imunologia , beta-Glucanas/farmacologia , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/imunologia , Diferenciação Celular/imunologia , Humanos , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos/citologia , Masculino , Monócitos/citologia , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/imunologia , beta-Glucanas/química
20.
Infect Immun ; 86(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30275010

RESUMO

The skin microenvironment at the site of infection plays a role in the early events that determine protective T helper 1/type 1 immune responses during cutaneous leishmaniasis (CL) infection. During CL in nonhealing BALB/c mice, early interleukin-4 (IL-4) can instruct dendritic cells for protective Th1 immunity. Additionally, keratinocytes, which are the principal cell type in the skin epidermis, have been shown to secrete IL-4 early after Leishmania major infection. Here, we investigated whether IL-4/IL-13 signaling via the common IL-4 receptor alpha chain (IL-4Rα) on keratinocytes contributes to susceptibility during experimental CL. To address this, keratinocyte-specific IL-4Rα-deficient (KRT14cre IL-4Rα-/lox) mice on a BALB/c genetic background were generated by gene targeting and site-specific recombination (Cre/loxP) under the control of the keratinocyte-specific krt14 locus. Following high-dose infection with L. major IL-81 and LV39 promastigotes subcutaneously in the footpad, footpad swelling, parasite burden, IFN-γ/IL-4/IL-13 cytokine production, and type 1 and type 2 antibody responses were similar between KRT14cre IL-4Rα-/lox and littermate control IL-4Rα-/lox BALB/c mice. An intradermal infection with low-dose L. major IL-81 and LV39 promastigotes in the ear showed results in infected KRT14cre IL-4Rα-/lox BALB/c mice similar to those of littermate control IL-4Rα-/lox BALB/c mice, with the exception of a significant decrease observed in parasite burden only at the site of LV39 infection in the ear. Collectively, our results show that autocrine and paracrine signaling of IL-4/IL-13 through the IL-4Rα chain on keratinocytes does not influence the establishment of a nonhealing Th2 immune response in BALB/c mice during L. major infection.


Assuntos
Deleção de Genes , Subunidade alfa de Receptor de Interleucina-4/genética , Queratinócitos/imunologia , Leishmaniose Cutânea/imunologia , Transdução de Sinais/imunologia , Animais , Comunicação Autócrina/imunologia , Linfócitos T CD4-Positivos , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/parasitologia , Feminino , Interleucina-13/imunologia , Queratinócitos/parasitologia , Leishmania major/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Comunicação Parácrina/imunologia , Células Th2/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA