Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 500(3): 564-568, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29679568

RESUMO

We show here that increased S100A8 and S100A9 protein expression is induced in spleen of animals with active inflammation or with inoculated tumors. In tumor bearing animals an increased expression was also detected in the lung. To further analyze the induced proteins, we performed chemical cross-linking followed by Western blotting. We observed in protein extracts from spleen that both S100A8/S100A9 heterodimers as well as S100A9 homodimers were formed, both after tumor and inflammatory challenge. The cellular source for S100A9 homodimers were CD11b+GR1+ cells. S100A9 homodimers were also secreted into the extracellular space. Lastly, in the spleen from normal and tumor bearing animals cells expressing relatively higher levels of S100A9 compared to S100A8 could be observed by immunohistochemistry. Taken together, these data show that the biologically potent dimeric form of S100A9 is induced in vivo in situations of tumor burden or inflammatory challenge.


Assuntos
Calgranulina B/metabolismo , Multimerização Proteica , Animais , Calgranulina A/metabolismo , Espaço Extracelular/metabolismo , Inflamação/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Baço/metabolismo
2.
BMC Cancer ; 16: 440, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27400708

RESUMO

BACKGROUND: Previous work has demonstrated immunomodulatory, anti-tumor, anti-metastatic and anti-angiogenic effects of the small molecule quinoline-3-carboxamide tasquinimod in pre-clinical cancer models. To better understand the anti-tumor effects of tasquinimod in transplantable tumor models, we have evaluated the impact of the compound both on recruitment of myeloid cells to tumor tissue and on tumor-induced myeloid cell expansion as these cells are known to promote tumor development. METHODS: Mice bearing subcutaneous 4 T1 mammary carcinoma tumors were treated with tasquinimod in the drinking water. A BrdU-based flow cytometry assay was utilized to assess the impact of short-term tasquinimod treatment on myeloid cell recruitment to tumors. Additionally, long-term treatment was performed to study the anti-tumor effect of tasquinimod as well as its effects on splenic myeloid cells and their progenitors. Myeloid cell populations were also immune-depleted by in vivo antibody treatment. RESULTS: Short-term tasquinimod treatment did not influence the proliferation of splenic Ly6C(hi) and Ly6G(hi) cells, but instead reduced the influx of Ly6C(hi) cells to the tumor. Treatment with tasquinimod for various periods of time after tumor inoculation revealed that the anti-tumor effect of this compound mainly operated during the first few days of tumor growth. Similar to tasquinimod treatment, antibody-mediated depletion of Ly6C(hi) cells within that same time frame, caused reduced tumor growth, thereby confirming a significant role for these cells in tumor development. Additionally, long-term tasquinimod treatment reduced the splenomegaly and expansion of splenic myeloid cells during a later phase of tumor development. In this phase, tasquinimod normalized the tumor-induced alterations in myeloerythroid progenitor cells in the spleen but had only limited impact on the same populations in the bone marrow. CONCLUSIONS: Our results indicate that tasquinimod treatment reduces tumor growth by operating early after tumor inoculation and that this effect is at least partially caused by reduced recruitment of Ly6C(hi) cells to tumor tissue. Long-term treatment also reduces the number of splenic myeloid cells and myeloerythroid progenitors, but these effects did not influence established rapidly growing tumors.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Quinolinas/farmacologia , Quinolonas/farmacologia , Administração Oral , Inibidores da Angiogênese/administração & dosagem , Animais , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Células Mieloides/patologia , Células Progenitoras Mieloides/efeitos dos fármacos , Mielopoese/efeitos dos fármacos , Quinolinas/administração & dosagem , Quinolonas/administração & dosagem , Baço/citologia , Baço/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Am J Pathol ; 182(5): 1671-80, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23506849

RESUMO

Quinoline-3-carboxamide compounds (Q compounds) have demonstrated efficacy in treating autoimmune disease in both humans and mice. However, the mode of action of these compounds is poorly understood. Here, we show that preventive treatment with the Q compound paquinimod (ABR-215757) during the first 5 days after induction of experimental autoimmune encephalomyelitis is sufficient to significantly ameliorate disease symptoms. Parallel cell-depletion experiments demonstrated that Ly6C(hi) inflammatory monocytes play an essential role in this phase. The paquinimod-induced amelioration correlated with reduced priming of antigen-specific CD4(+) T cells and reduced frequency of IFN-γ- and IL-17-producing cells in draining lymph nodes. Importantly, the treatment did not inhibit T-cell division per se. In mice with established experimental autoimmune encephalomyelitis, the numbers of Ly6C(hi) CD115(+) inflammatory monocytes and CD11b(+)CD11c(+) dendritic cells (DCs) were reduced in spleen, but not in bone marrow or draining lymph nodes of treated mice. Inflammatory monocyte-derived DCs and CD4(+) T cells were also reduced in the brain. In contrast, there was no decrease in DC subsets previously shown to be critical for effector CD4(+) T-cell development in lymph nodes. Taken together, these data indicate that preventive treatment with paquinimod ameliorates experimental autoimmune encephalomyelitis by reducing effector T-cell priming and, on prolonged treatment, displays a selective effect by decreasing distinct subpopulations of splenic CD11b(+) myeloid cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Apresentação Cruzada/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Inflamação/patologia , Quinolinas/uso terapêutico , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Apresentação Cruzada/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Memória Imunológica/efeitos dos fármacos , Inflamação/imunologia , Interferon gama/biossíntese , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Quinolinas/farmacologia , Baço/patologia
4.
Rheumatology (Oxford) ; 52(11): 2048-55, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23942785

RESUMO

OBJECTIVES: Patients with SLE have an increased morbidity and mortality from cardiovascular disease (CVD). The reason for this is not entirely understood, but is believed to be partly related to the long-lasting inflammatory process seen in SLE. The aim of the present study was to investigate whether there is an association between CVD and serum levels of the proinflammatory proteins S100A8/A9 and S100A12 in SLE. METHODS: Serum levels of S100A8/A9 and S100A12 were measured with ELISA in 237 SLE patients with clinically inactive disease and without infections, as well as in 100 healthy individuals. Cardiovascular manifestations were defined according to the SLICC/ACR Damage Index (SLICC/ACR-DI). RESULTS: Serum levels of S100A8/A9 were elevated in our inactive SLE patients as compared with healthy individuals (P < 0.0001), which was not seen for S100A12 (P = 0.12). SLE patients with a history of CVD had increased serum levels of both S100A8/A9 and S100A12 compared with patients with no CVD or venous thromboembolism (P = 0.003 and P = 0.006, respectively). The presence of organ damage according to SLICC/ACR-DI was associated with an increase in both S100A8/A9 and S100A12 serum levels (P = 0.001 and P = 0.006, respectively). CONCLUSION: Elevated serum levels of S100A8/A9 and S100A12 may be used as an indicator of severe disease and CVD in SLE, suggesting that SLE patients with elevated serum S100A8/A9 and S100A12 concentrations may benefit from more intense cardiovascular primary preventive strategies and possibly also from more intense and early immunosuppressive treatment.


Assuntos
Doenças Cardiovasculares/etiologia , Lúpus Eritematoso Sistêmico/complicações , Proteínas S100/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Calgranulina A/sangue , Calgranulina B/sangue , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/diagnóstico , Estudos de Casos e Controles , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Lúpus Eritematoso Sistêmico/sangue , Masculino , Pessoa de Meia-Idade , Proteína S100A12 , Índice de Gravidade de Doença , Adulto Jovem
5.
Immunology ; 137(2): 172-82, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22804476

RESUMO

Interactions between danger-associated molecular patterns (DAMP) and pathogen-associated molecular patterns (PAMP) and pattern recognition receptors such as Toll-like receptors (TLRs) are critical for the regulation of the inflammatory process via activation of nuclear factor-κB (NF-κB) and cytokine secretion. In this report, we investigated the capacity of lipopolysaccharide (LPS) -free S100A9 (DAMP) protein to activate human and mouse cells compared with lipoprotein-free LPS (PAMP). First, we showed that LPS and S100A9 were able to increase NF-κB activity followed by increased cytokine and nitric oxide (NO) secretion both in human THP-1 cells and in mouse bone marrow-derived dendritic cells. Surprisingly, although S100A9 triggered a weaker cytokine response than LPS, we found that S100A9 more potently induced IκBα degradation and hence NF-κB activation. Both the S100A9-induced response and the LPS-induced response were completely absent in TLR4 knockout mice, whereas it was only slightly affected in RAGE knockout mice. Also, we showed that LPS and S100A9 NF-κB induction were strongly reduced in the presence of specific inhibitors of TLR-signalling. Chloroquine reduced S100A9 but not LPS signalling, indicating that S100A9 may need to be internalized to be fully active as a TLR4 inducer. This was confirmed using A488-labelled S100A9 that was internalized in THP-1 cells, showing a raise in fluorescence after 30 min at 37°. Chloroquine treatment significantly reduced the fluorescence. In summary, our data indicate that both human and mouse S100A9 are TLR4 agonists. Importantly, S100A9 induced stronger NF-κB activation albeit weaker cytokine secretion than LPS, suggesting that S100A9 and LPS activated NF-κB in a qualitatively distinct manner.


Assuntos
Calgranulina B/imunologia , NF-kappa B/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/deficiência
6.
J Exp Med ; 203(4): 1007-19, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16585265

RESUMO

The endothelial cell monolayer of cerebral vessels and its basement membrane (BM) are ensheathed by the astrocyte endfeet, the leptomeningeal cells, and their associated parenchymal BM, all of which contribute to establishment of the blood-brain barrier (BBB). As a consequence of this unique structure, leukocyte penetration of cerebral vessels is a multistep event. In mouse experimental autoimmune encephalomyelitis (EAE), a widely used central nervous system inflammatory model, leukocytes first penetrate the endothelial cell monolayer and underlying BM using integrin beta1-mediated processes, but mechanisms used to penetrate the second barrier defined by the parenchymal BM and glia limitans remain uninvestigated. We show here that macrophage-derived gelatinase (matrix metalloproteinase [MMP]-2 and MMP-9) activity is crucial for leukocyte penetration of the parenchymal BM. Dystroglycan, a transmembrane receptor that anchors astrocyte endfeet to the parenchymal BM via high affinity interactions with laminins 1 and 2, perlecan and agrin, is identified as a specific substrate of MMP-2 and MMP-9. Ablation of both MMP-2 and MMP-9 in double knockout mice confers resistance to EAE by inhibiting dystroglycan cleavage and preventing leukocyte infiltration. This is the first description of selective in situ proteolytic damage of a BBB-specific molecule at sites of leukocyte infiltration.


Assuntos
Membrana Basal/metabolismo , Distroglicanas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Migração e Rolagem de Leucócitos/imunologia , Animais , Astrócitos/metabolismo , Membrana Basal/enzimologia , Células Cultivadas , Encefalomielite Autoimune Experimental/enzimologia , Gelatinases/genética , Gelatinases/metabolismo , Hidrólise , Leucócitos/citologia , Leucócitos/enzimologia , Macrófagos/enzimologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
BMC Immunol ; 13: 69, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23234398

RESUMO

BACKGROUND: S100A9 has been shown to be important for the function of so called Myeloid Derived Suppressor Cells (MDSC). Cells with a similar phenotype are also involved in pro-inflammatory processes, and we therefore wanted to investigate the gene expression and function of these cells in animals that were either subjected to chronic inflammation, or inoculated with tumors. METHODS: CD11b(+)Ly6C(++) and Ly6G(+) cells were isolated from spleen, tumor tissue or inflammatory granulomas. S100A9, Arginase 1 and iNOS gene expression in the various CD11b(+) cell populations was analyzed using Q-PCR. The suppressive activity of the CD11b(+) cell populations from different donors was studied in co-culture experiments. RESULTS: S100A9 was shown to be expressed mainly in splenic CD11b(+)Ly6C(+)G(+) cells both at the RNA and protein level. Arginase I and iNOS expression could be detected in both CD11b(+)Ly6C(+)Ly6G(+) and CD11b(+)Ly6C(+)G(-)/C(++)G(-) derived from tumors or a site of chronic inflammation, but was very low in the same cell populations isolated from the spleen. CD11b(+) cells isolated from mice with peritoneal chronic inflammation were able to stimulate T lymphocytes, while CD11b+ cells from mice with peritoneal tumors suppressed T cell growth. CONCLUSION: An identical CD11b(+)Ly6C(++)G(-) cell population appears to have the ability to adopt immune stimulatory or immune suppressive functions dependent on the presence of a local inflammatory or tumor microenvironment. Thus, there is a functional plasticity in the CD11b(+)Ly6C(++)G(-) cell population that cannot be distinguished with the current molecular markers.


Assuntos
Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Inflamação/imunologia , Neoplasias/imunologia , Carga Tumoral/imunologia , Animais , Arginase/metabolismo , Calgranulina B/metabolismo , Proliferação de Células , Doença Crônica , Feminino , Inflamação/enzimologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/enzimologia , Neoplasias/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Baço/metabolismo , Baço/patologia , Linfócitos T/citologia , Linfócitos T/imunologia , Terpenos , Microambiente Tumoral/imunologia
8.
PLoS Biol ; 7(4): e97, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19402754

RESUMO

Despite more than 25 years of research, the molecular targets of quinoline-3-carboxamides have been elusive although these compounds are currently in Phase II and III development for treatment of autoimmune/inflammatory diseases in humans. Using photoaffinity cross-linking of a radioactively labelled quinoline-3-carboxamide compound, we could determine a direct association between human S100A9 and quinoline-3-carboxamides. This interaction was strictly dependent on both Zn++ and Ca++. We also show that S100A9 in the presence of Zn++ and Ca++ is an efficient ligand of receptor for advanced glycation end products (RAGE) and also an endogenous Toll ligand in that it shows a highly specific interaction with TLR4/MD2. Both these interactions are inhibited by quinoline-3-carboxamides. A clear structure-activity relationship (SAR) emerged with regard to the binding of quinoline-3-carboxamides to S100A9, as well as these compounds potency to inhibit interactions with RAGE or TLR4/MD2. The same SAR was observed when the compound's ability to inhibit acute experimental autoimmune encephalomyelitis in mice in vivo was analysed. Quinoline-3-carboxamides would also inhibit TNFalpha release in a S100A9-dependent model in vivo, as would antibodies raised against the quinoline-3-carboxamide-binding domain of S100A9. Thus, S100A9 appears to be a focal molecule in the control of autoimmune disease via its interactions with proinflammatory mediators. The specific binding of quinoline-3-carboxamides to S100A9 explains the immunomodulatory activity of this class of compounds and defines S100A9 as a novel target for treatment of human autoimmune diseases.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Calgranulina B/metabolismo , Fatores Imunológicos/farmacologia , Inflamação/metabolismo , Quinolinas/metabolismo , Receptores Imunológicos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Doenças Autoimunes/metabolismo , Cálcio/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/efeitos adversos , Antígeno 96 de Linfócito/metabolismo , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Quinolinas/química , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Zinco/metabolismo
10.
Prostate ; 70(13): 1461-70, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20687219

RESUMO

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) activity has been shown to be expressed in local lymph nodes and induce immune suppression of tumor immunity. Here we analyze the effect of IDO expression on prostate tumor growth using the transgenic adenocarcinoma of mouse prostate (TRAMP) animal model. METHODS: Mice deficient in IDO expression were crossed to TRAMP mice and the time to the appearance of palpable tumors were measured. Immune histology was used to analyze the IDO expressing cells in tumors and in local lymph nodes. The levels of the substrate for IDO (tryptophane) and its product (kynurenine) was measured by HPLC. RESULTS: We found that systemic IDO activity, determined as the kynurenine/tryptophan ratio in serum, correlated with the presence of palpable tumor. Immunohistological analysis showed increased numbers of IDO expressing cells in local lymph nodes. In tumors, IDO expression could be detected in the tumor stroma by both CD31+ and CD31(-) cells. Essentially no CD45+, IDO expressing cells could be detected in the tumors. The influence of IDO activity on tumor progression was analyzed by back-crossing TRAMP mice with IDO(-/-) animals and J-chain negative (J(-/-)) mice that have perturbed IDO activity. In both crosses a delayed tumor incidence was observed. CONCLUSION: Our results argue for a role for IDO mediated immune suppression in the early stages of prostate cancer progression. However, since the intra-tumor IDO expression in J(-/-) mice was indistinguishable from that of C57BL/6 animals the IDO expression in the tumor tissue appears to be irrelevant for TRAMP tumor incidence.


Assuntos
Adenocarcinoma/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Linfonodos/metabolismo , Neoplasias da Próstata/metabolismo , Adenocarcinoma/patologia , Animais , Western Blotting , Progressão da Doença , Imuno-Histoquímica , Cinurenina/sangue , Linfonodos/patologia , Masculino , Camundongos , Neoplasias da Próstata/patologia , Triptofano/sangue
11.
Eur J Immunol ; 39(4): 1078-87, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19283777

RESUMO

Repeated immunization of mice with bacterial superantigens induces extensive deletion and anergy of reactive CD4 T cells. Here we report that the in vitro proliferation anergy of CD4 T cells from TCR transgenic mice immunized three times with staphylococcal enterotoxin B (SEB) (3 x SEB) is partially due to an increased frequency of Foxp3(+) CD4 T cells. Importantly, reduced number of conventional CD25(-) Foxp3(-) cells, rather than conversion of such cells to Foxp3(+) cells, was the cause of that increase and was also seen in mice repeatedly immunized with OVA (3 x OVA) and OVA-peptide (OVAp) (3 x OVAp). Cell-transfer experiments revealed profound but transient anergy of CD4 T cells isolated from 3 x OVAp and 3x SEB mice. However, the in vivo anergy was CD4 T-cell autonomous and independent of Foxp3(+) Treg. Finally, proliferation of transferred CD4 T cells was inhibited in repeatedly immunized mice but inhibition was lost when transfer was delayed, despite the maintenance of elevated frequency of Foxp3(+) cells. These data provide important implications for Foxp3(+) cell-mediated tolerance in situations of repeated antigen exposure such as human persistent infections.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Anergia Clonal/imunologia , Fatores de Transcrição Forkhead/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proliferação de Células , Enterotoxinas/imunologia , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Superantígenos/imunologia
12.
Commun Biol ; 2: 176, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31098409

RESUMO

Innate immune responses are rapid, dynamic and highly regulated to avoid overt reactions. This regulation is executed by innate immune tolerance mechanisms that remain obscure. Wnt5a is a signalling protein mainly involved in developmental processes and cancer. The effect of Wnt5a on inflammatory myeloid cells is controversial. Here, we combine primary cell cultures, in vitro binding studies, mass spectrometry and Drosophila protein modelling to show that Wnt5a is a direct ligand of toll-like receptor (TLR) 2 and 4. The binding promotes a MyD88-non-canonical nuclear factor of kappa B (NFκB) and AP-1 signalling cascade, with contradictory profiles in mouse (pro-inflammatory) and human (anti-inflammatory) myeloid immune cells. These data reveal that the true nature of Wnt5a in inflammatory cells, is to regulate TLR signals, and in human myeloid cells it acts as an endogenous, tolerance-associated molecular pattern (TAMP), inducing IL-10 and innate immune tolerance.


Assuntos
Células Mieloides/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Proteína Wnt-5a/imunologia , Animais , Células Cultivadas , Citocinas/biossíntese , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Humanos , Tolerância Imunológica , Imunidade Inata , Interleucina-10/biossíntese , Interleucina-10/genética , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Camundongos , Modelos Imunológicos , Modelos Moleculares , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptores Toll-Like/química , Receptores Toll-Like/metabolismo , Transcrição Gênica , Proteína Wnt-5a/metabolismo
13.
Sci Signal ; 12(596)2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31455723

RESUMO

Second mitochondria-derived activator of caspase (SMAC) mimetics (SMs) are selective antagonists of the inhibitor of apoptosis proteins (IAPs), which activate noncanonical NF-κB signaling and promote tumor cell death. Through gene expression analysis, we found that treatment of CD4+ T cells with SMs during T helper 17 (TH17) cell differentiation disrupted the balance between two antagonistic transcription factor modules. Moreover, proteomics analysis revealed that SMs altered the abundance of proteins associated with cell cycle, mitochondrial activity, and the balance between canonical and noncanonical NF-κB signaling. Whereas SMs inhibited interleukin-17 (IL-17) production and ameliorated TH17 cell-driven inflammation, they stimulated IL-22 secretion. Mechanistically, SM-mediated activation of NF-κB-inducing kinase (NIK) and the transcription factors RelB and p52 directly suppressed Il17a expression and IL-17A protein production, as well as the expression of a number of other immune genes. Induction of IL-22 production correlated with the NIK-dependent reduction in cMAF protein abundance and the enhanced activity of the aryl hydrocarbon receptor. Last, SMs also increased IL-9 and IL-13 production and, under competing conditions, favored the differentiation of naïve CD4+ T cells into TH2 cells rather than TH17 cells. These results demonstrate that SMs shape the gene expression and protein profiles of TH17 cells and inhibit TH17 cell-driven autoimmunity.


Assuntos
Proteínas Reguladoras de Apoptose , Materiais Biomiméticos/farmacologia , Diferenciação Celular/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Mitocondriais , Proteínas Serina-Treonina Quinases/imunologia , Células Th17/imunologia , Animais , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Transgênicos , Células Th17/citologia , Células Th2/citologia , Células Th2/imunologia , Quinase Induzida por NF-kappaB
14.
PLoS One ; 13(5): e0196598, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29742113

RESUMO

Quinoline-3-carboxamides (Q compounds) are immunomodulatory compounds that have shown efficacy both in autoimmune disease and cancer. We have in here investigated the impact of one such compound, paquinimod, on the development of diabetes in the NOD mouse model for type I diabetes (T1D). In cohorts of NOD mice treated with paquinimod between weeks 10 to 20 of age and followed up until 40 weeks of age, we observed dose-dependent reduction in incidence of disease as well as delayed onset of disease. Further, in contrast to untreated controls, the majority of NOD mice treated from 15 weeks of age did not develop diabetes at 30 weeks of age. Importantly, these mice displayed significantly less insulitis, which correlated with selectively reduced number of splenic macrophages and splenic Ly6Chi inflammatory monocytes at end point as compared to untreated controls. Collectively, these results demonstrate that paquinimod treatment can significantly inhibit progression of insulitis to T1D in the NOD mouse. We propose that the effect of paquinimod on disease progression may be related to the reduced number of these myeloid cell populations. Our finding also indicates that this compound could be a candidate for clinical development towards diabetes therapy in humans.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Imunossupressores/uso terapêutico , Quinolinas/uso terapêutico , Animais , Diabetes Mellitus Tipo 1/patologia , Feminino , Glicosúria/prevenção & controle , Imunossupressores/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Monócitos/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Quinolinas/farmacologia
15.
PLoS One ; 13(9): e0203228, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30183741

RESUMO

Quinoline-3-carboxamides (Q substances) are small molecule compounds with anti-inflammatory properties. In this study, we used one of these substances, Paquinimod, to treat a novel model for chronic liver inflammation and liver fibrosis, the NOD-Inflammation Fibrosis (N-IF) mouse. We show that treatment of N-IF mice significantly reduced inflammation and resulted in the regression of fibrosis, even when the treatment was initiated after onset of disease. The reduced disease phenotype was associated with a systemic decrease in the number and reduced activation of disease-promoting transgenic natural killer T (NKT)-II cells and their type 2-cytokine expression profile. Paquinimod treatment also led to a reduction of CD115+ Ly6Chi monocytes and CD11b+ F4/80+ CD206+ macrophages.


Assuntos
Fatores Imunológicos/farmacologia , Cirrose Hepática/tratamento farmacológico , Quinolinas/farmacologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/patologia , Células T Matadoras Naturais/efeitos dos fármacos , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/patologia
16.
Chem Immunol Allergy ; 93: 137-160, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17369704

RESUMO

Bacterial and viral superantigens (SAgs) can induce the activation of a large fraction of the T cells in an individual. The activated T cells divide and produce a strong cytokine response. Subsequently, the majority of the expanded T cells are deleted and the remaining SAg-reactive cells will be anergic and tolerant to SAg exposure. The anergic population is heterogeneous and includes both T cells with induced nonresponsiveness to the SAg and SAg-specific regulatory T cells (Tregs). Both CD4+CD25- and CD4+CD25+ Tregs are generated in SAg-induced responses. The Tregs suppress T cell activation and produce the immunosuppressive cytokines IL-10 and TGF-Beta1, thereby contributing to the SAg-specific tolerance. The similarities of immune responses induced by SAgs and conventional peptide antigens administered in the absence of adjuvants will be discussed. It is proposed that the antigen-specific tolerance induced in both situations is the consequence of T cell activation mediated by immature dendritic cells.


Assuntos
Superantígenos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anergia Clonal , Citocinas/biossíntese , Humanos , Interleucina-10/biossíntese
17.
Transplantation ; 82(11): 1493-500, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17164722

RESUMO

BACKGROUND: Previous studies have demonstrated that anti-CD40L or anti-B7 requires the presence of CD4(+)CD25(+) regulatory T cells (Treg) to induce antigen specific hyporesponsiveness. Other tolerance strategies involving Treg have shown a dependency on interleukin (IL)-10. The objective of this study was to investigate the role of CD4(+)CD25(+) Treg and IL-10 when treating transplant recipients with cytotoxic T lymphocyte-associated antigen (CTLA)-4 immunoglobulin (Ig), anti-CD40L, and anti-lymphocyte function-associated antigen (LFA)-1. METHODS: Recombinase activating gene-deficient (Rag1(-/-) mice were transplanted with BALB/c hearts and adoptively transferred with IL-10(-/-) CD4(+) T cells, CD4(+)CD25(-) T cells or CD4(+)CD25(-)CD103(-) T cells and treated with costimulation blockade. Intragraft T cells from C57BL/6 recipients were analyzed for the expression of the Foxp3 protein after tolerance induction. RESULTS: Mice reconstituted with IL-10(-/-) CD4(+) T cells, CD4(+)CD25(-) T cells or CD4(+)CD25(-) CD103(-) T cells and treated with costimulation blockade accepted allografts permanently. Analysis of cells from recipient mice adoptively transferred with CD4(+)CD25(-) T cells contained a population of CD4(low)CD25(+) T cells 100 days after transplantation. Costimulation blockade partially prevented the homeostatic proliferation of CD4(+)CD25(-)CD103(-) T cells in Rag-1(-/-) recipients. Accepted allografts contained an elevated number of CD4(+)Foxp3(+) T cells. CONCLUSIONS: These results indicate that T-cell derived IL-10 is not essential for induction of graft acceptance in mice treated with costimulation blockade, but that treatment limits T-cell expansion in the recipients. The results further indicate that tolerance is maintained by intragraft CD4(+)Foxp3(+) T cells.


Assuntos
Antígenos CD/farmacologia , Antígenos de Diferenciação/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Coração/imunologia , Imunoglobulina G/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Tolerância ao Transplante , Animais , Antígenos CD4/análise , Ligante de CD40/antagonistas & inibidores , Ligante de CD40/imunologia , Antígeno CTLA-4 , Feminino , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/metabolismo , Genes RAG-1/genética , Sobrevivência de Enxerto/imunologia , Interleucina-10/genética , Subunidade alfa de Receptor de Interleucina-2/análise , Antígeno-1 Associado à Função Linfocitária/efeitos dos fármacos , Antígeno-1 Associado à Função Linfocitária/imunologia , Camundongos , Camundongos Mutantes , Transplante de Pele , Linfócitos T Reguladores/imunologia
18.
PLoS One ; 11(5): e0156377, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27228163

RESUMO

The cytosolic Ca2+-binding S100A9 and S100A8 proteins form heterodimers that are primarily expressed in human neutrophils and monocytes. We have recently shown that S100A9 binds to TLR4 in vitro and induces TLR4-dependent NF-κB activation and a pro-inflammatory cytokine response in monocytes. In the present report we have further investigated the S100A9-mediated stimulation of TLR4 in monocytes. Using transmission immunoelectron microscopy, we detected focal binding of S100A9 to monocyte membrane subdomains containing the caveolin-1 protein and TLR4. Furthermore, the S100A9 protein was detected in early endosomes of the stimulated cells, indicating that the protein could be internalized by endocytosis. Although stimulation of monocytes with S100A9 was strictly TLR4-dependent, binding of S100A9 to the plasma membrane and endocytosis of S100A9 was still detectable and coincided with CD14 expression in TLR4-deficient cells. We therefore investigated whether CD14 would be involved in the TLR4-dependent stimulation and could show that the S100A9-induced cytokine response was inhibited both in CD14-deficient cells and in cells exposed to CD14 blocking antibodies. Further, S100A9 was not internalized into CD14-deficient cells suggesting a direct role of CD14 in endocytosis of S100A9. Finally, we could detect satiable binding of S100A9 to CD14 in surface plasmon resonance experiments. Taken together, these results indicate that CD14 is a co-receptor of TLR4 in the S100A9-induced cytokine response.


Assuntos
Calgranulina B/imunologia , Receptores de Lipopolissacarídeos/imunologia , Monócitos/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Calgranulina B/genética , Caveolina 1/genética , Caveolina 1/imunologia , Linhagem Celular Tumoral , Membrana Celular/genética , Membrana Celular/imunologia , Citocinas/genética , Citocinas/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Receptores de Lipopolissacarídeos/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Ressonância de Plasmônio de Superfície , Receptor 4 Toll-Like/genética
19.
Arthritis Rheumatol ; 68(8): 1970-80, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26946461

RESUMO

OBJECTIVE: Levels of S100A8/A9, a proinflammatory and prothrombotic protein complex, are increased in several diseases, and high levels predispose to cardiovascular disease (CVD). Recently, platelet S100A8/A9 synthesis was described in mice and humans in relation to CVD. The aim of this study was to investigate the role of platelet S100A8/A9 in systemic lupus erythematosus (SLE), a disease with markedly increased cardiovascular morbidity, as well as the exact platelet distribution of the S100A8/A9 proteins. METHODS: The occurrence and distribution of platelet S100A8/A9 protein were detected by enzyme-linked immunosorbent assay, electron microscopy, Western blotting, and flow cytometry in healthy controls (n = 79) and in 2 individual cohorts of SLE patients (n = 148 and n = 318, respectively) and related to cardiovascular morbidity. RESULTS: We observed that human platelets expressed S100A8/A9 proteins, and that these were localized in close proximity to intracellular membranes and granules as well as on the cell surface upon activation with physiologic and pathophysiologic stimuli. Interestingly, S100A8/A9 was enriched at sites of membrane interactions, indicating a role of S100A8/A9 in cell-cell communication. S100A8/A9 levels were highly regulated by interferon-α, both in vivo and in vitro. Patients with SLE had increased platelet S100A8/A9 content compared with healthy individuals. Increased levels of platelet S100A8/A9 were associated with CVD, particularly myocardial infarction (odds ratio 4.8, 95% confidence interval 1.5-14.9, P = 0.032 [adjusted for age, sex, and smoking]). CONCLUSION: Platelets contain S100A8/A9 in membrane-enclosed vesicles, enabling rapid cell surface deposition upon activation. Furthermore, platelet S100A8/A9 protein levels were increased in SLE patients, particularly in those with CVD, and may be a future therapeutic target.


Assuntos
Plaquetas , Calgranulina A/sangue , Calgranulina B/sangue , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/etiologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/complicações , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Plaquetas/metabolismo , Calgranulina A/biossíntese , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
20.
PLoS One ; 11(7): e0159850, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27441847

RESUMO

Here we characterize a new animal model that spontaneously develops chronic inflammation and fibrosis in multiple organs, the non-obese diabetic inflammation and fibrosis (N-IF) mouse. In the liver, the N-IF mouse displays inflammation and fibrosis particularly evident around portal tracts and central veins and accompanied with evidence of abnormal intrahepatic bile ducts. The extensive cellular infiltration consists mainly of macrophages, granulocytes, particularly eosinophils, and mast cells. This inflammatory syndrome is mediated by a transgenic population of natural killer T cells (NKT) induced in an immunodeficient NOD genetic background. The disease is transferrable to immunodeficient recipients, while polyclonal T cells from unaffected syngeneic donors can inhibit the disease phenotype. Because of the fibrotic component, early on-set, spontaneous nature and reproducibility, this novel mouse model provides a unique tool to gain further insight into the underlying mechanisms mediating transformation of chronic inflammation into fibrosis and to evaluate intervention protocols for treating conditions of fibrotic disorders.


Assuntos
Hepatite Crônica/etiologia , Hepatite Crônica/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Transferência Adotiva , Animais , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Biomarcadores , Citocinas/metabolismo , Modelos Animais de Doenças , Hepatite Crônica/metabolismo , Mediadores da Inflamação/metabolismo , Cirrose Hepática/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Fenótipo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA