Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Biomed Res Int ; 2021: 9975865, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34708129

RESUMO

Interleukin-18 (IL-18) is an inflammatory cytokine that has been linked to energy homeostasis and psychiatric symptoms such as depression and cognitive impairment. We previously revealed that deficiency in IL-18 led to hippocampal abnormalities and resulted in depression-like symptoms. However, the impact of IL-18 deficiency on other brain regions remains to be clarified. In this study, we first sought to confirm that IL-18 expression in neural cells can be found in human brain tissue. Subsequently, we examined the expression of genes in the prefrontal cortex of Il18 -/- mice and compared it with gene expression in mice subjected to a chronic mild stress model of depression. Extracted genes were further analyzed using Ingenuity® Pathway Analysis, in which 18 genes common to both the chronic mild stressed model and Il18 -/- mice were identified. Of those, 16 were significantly differentially expressed between Il18+/+ and Il18 -/- mice. We additionally measured protein expression of α-2-HS-glycoprotein (AHSG) and transthyretin (TTR) in serum and the brain. In the prefrontal cortex of Il18 -/- mice, TTR but not AHSG was significantly decreased. Conversely, in the serum of Il18 -/- mice, AHSG was significantly increased but not TTR. Therefore, our results suggest that in IL-18-deficit conditions, TTR in the brain is one of the mediators causally related to depression, and AHSG in peripheral organs is one of the regulators inducing energy imbalance. Moreover, this study suggests a possible "signpost" to clarify the molecular mechanisms commonly underlying the immune system, energy metabolism, neural function, and depressive disorders.


Assuntos
Transtorno Depressivo/imunologia , Interleucina-18/deficiência , Interleucina-18/metabolismo , Adulto , Animais , Encéfalo/metabolismo , Depressão/imunologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo
2.
Nutrients ; 12(11)2020 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-33121026

RESUMO

Lactic acid bacteria (LAB), a major commensal bacterium in the small intestine, are well known beneficial bacteria which promote establishment of gut-centric immunity, such as anti-inflammation and anti-infection. In this report, we show that a LAB strain Lactococcus lactis subsp. Cremoris C60 possess an ability to activate antigen presenting cells, such as dendritic cells (DCs), and intestinal T cells which possibly support to maintain healthy intestinal immunological environment in aging process. We found that CD4+ T cells in the small intestine are dramatically decreased in aged Interleukin-18 knock out (IL-18KO) mice, associated with the impairment of IFN-γ production in the CD4+ T cells, especially in small intestinal lamina propria (LP). Surprisingly, heat killed-C60 (HK-C60) diet completely recovered the CD4+ T cells population and activity in SI-LP and over activated the population in Peyer's patches (PPs) of IL-18KO mice. The HK-C60 diet was effective approach not only to restore the number of cells, but also to recover IFN-γ production in the CD4+ T cell population in the small intestine of IL-18-deficient mice. As a possible cause in the age-associated impairment of CD4+ T cells activity in IL-18KO mice, we found that the immunological activity was downregulated in the IL-18-deficient DCs. The cytokines production and cellular activation markers expression were downregulated in the IL-18-deficient bone marrow derived dendritic cells (BMDCs) at the basal level, however, both activities were highly upregulated in HK-C60 stimulation as compared to those of WT cells. Antigen uptake was also attenuated in the IL-18-deficient BMDCs, and it was significantly enhanced in the cells as compared to WT cells in HK-60 stimulation. An in vitro antigen presentation assay showed that IFN-γ production in the CD4+ T cells was significantly enhanced in the culture of IL-18-deficient BMDCs compared with WT cells in the presence of HK-C60. Thus, we conclude that HK-C60 diet possesses an ability to restore T cells impairment in the small intestine of IL-18-deficient environment. In addition, the positive effect is based on the immunological modification of DCs function which directory influences into the promotion of effector CD4+ T cells generation in the small intestine.


Assuntos
Interleucina-18/deficiência , Intestino Delgado/imunologia , Lactococcus/fisiologia , Probióticos/administração & dosagem , Linfócitos T/imunologia , Envelhecimento , Animais , Linfócitos T CD4-Positivos , Contagem de Células , Células Dendríticas/imunologia , Dieta , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa/imunologia , Linfócitos T/citologia
3.
Am J Physiol Heart Circ Physiol ; 317(5): H1142-H1156, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31518161

RESUMO

IL-18 is ubiquitously produced by both hematopoietic and non-hematopoietic cells. The present study examined the thoracic aorta, including the surrounding perivascular adipose tissue (PVAT), of IL-18KO mice from functional and histological perspectives. IL-18KO mice exhibited raised blood pressure compared with wild-type mice. Echocardiographic examination showed a thickened vascular wall and narrowed vascular diameter of the aorta. Examination by the Magnus test demonstrated dysfunction of endothelial cells (ECs) in the IL-18KO thoracic aorta and impairment of the anticontractile function of IL-18KO PVAT. Histological examination showed no inflammatory lesions in the aorta but indicated progressive fibrosis in the vessel and conversion of PVAT from brown adipose tissue-like features to white adipose tissue-like features. Electron microscopic observation suggested several deformed mitochondria in the aorta and vacuole-like structures in ECs from IL-18KO mice. In addition, activity of complex IV was lower and production of reactive oxygen species was augmented in the mitochondria of IL-18KO aorta. Although expression of LC3 B was higher, rapamycin-induced autophagy flux was impaired in the IL-18KO PVAT. Moreover, Western blot analysis revealed that LAMP 1/2 was increased in IL-18KO PVAT, and measurement of cathepsin-D activity indicated decreased levels in IL-18KO PVAT. The IL-18KO thoracic aorta thus showed defects in physiological functions related to histological alterations, and the inflammasome/IL-18 system was suggested to play a protective role in cardiovascular cells, probably through quality control of mitochondria via promotion of autophagosome/autophagolysosome formation.NEW & NOTEWORTHY IL-18 deficiency caused aortic abnormalities in terms of morphology and functions in parallel with an accumulation of damaged mitochondria and anomalous turnover of protein complexes, such as PGC-1 and LAMP1 and -2 in PVAT. These findings suggested that IL-18 plays roles in maintaining the homeostasis of vessels and PVAT around the aorta, possibly by promoting autophagy.


Assuntos
Tecido Adiposo/metabolismo , Aorta Torácica/metabolismo , Autofagia , Interleucina-18/deficiência , Mitocôndrias/metabolismo , Tecido Adiposo/fisiopatologia , Tecido Adiposo/ultraestrutura , Animais , Aorta Torácica/fisiopatologia , Aorta Torácica/ultraestrutura , Metabolismo Energético , Hemodinâmica , Interleucina-18/genética , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
4.
J Transl Med ; 16(1): 314, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30453990

RESUMO

BACKGROUND: The cytokine, interleukin-18 (IL-18), was originally identified as an interferon-γ-inducing proinflammatory factor; however, there is increasing evidence suggesting that it has non-immunological effects on physiological functions. We have previously investigated the potential pathophysiological relationship between IL-18 and dyslipidemia, non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, which were mediated by lipid energy imbalance. Therefore, herein we focused on brown adipocytes (BAs) and brown adipose tissue (BAT) related to energy consumption as non-shivering thermogenesis. METHODS: Il18-/- male mice were generated on the C57Bl/6 background, and littermate C57Bl/6 Il18+/+ male mice were used as controls. To reveal the direct effect of IL-18, primary cell cultures derived from both mice were established. Moreover, for molecular analysis, microarray, quantitative reverse transcription PCR and western blotting were performed using 6 and 12 weeks old mice. To evaluate the short- and long-term effects of IL-18 on BAT, recombinant IL-18 was administered for 2 and 12 weeks, respectively. RESULTS: Compared with Il18+/+ mice, BAT of Il18-/- mice showed earlier differentiation and lipid accumulation. To examine the direct effect of IL-18 on BAT, BA cell cultures were established. Myogenic factor 5-expressing adipose precursor cells were extracted from Il18+/+ and Il18-/- mice. PR domain containing 16 (PRDM16), a differentiation inducer, was strongly expressed in Il18-/- BAs, and uncoupling protein 1, a thermogenic and differentiation marker, was upregulated, resulting in the promotion of BA differentiation. Moreover, PRDM16-dependent and independent molecules related to BAT function, such as fibroblast growth factor 21, were activated. These findings were confirmed by comparing Il18+/+ and Il18-/- mice at 6 and 12 weeks of age. Additional analyses of the molecular mechanisms influencing the 'Quantity of adipocytes' identified three associated genes, apolipoprotein C3 (Apoc3), insulin-induced gene 1 (Insig1) and vitamin D (1,25-dihydroxyvitamin D3) receptor (Vdr). Intravenous administration of IL-18 not only significantly improved the expression of some of these genes, but it also significantly decreased the adipocytes' size. CONCLUSIONS: This study demonstrated the critical function of IL-18 in differentiation and lipid metabolism in BAs. Furthermore, IL-18 may contribute to novel treatments by improving the energy imbalance.


Assuntos
Tecido Adiposo Marrom/patologia , Adiposidade , Diferenciação Celular , Dislipidemias/metabolismo , Dislipidemias/patologia , Interleucina-18/deficiência , Adipogenia/efeitos dos fármacos , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/crescimento & desenvolvimento , Animais , Diferenciação Celular/efeitos dos fármacos , Fígado Gorduroso/patologia , Interleucina-18/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Termogênese/efeitos dos fármacos
5.
Biochem Pharmacol ; 155: 150-161, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29963998

RESUMO

Interleukin (IL)-1 and IL-18 belong to the IL-1 family of ligands, and their receptors are members of the IL-1 receptor family. Both cytokines drive an extensive range of pro-inflammatory networks in many cell types using common signal transduction cascades. Anyway, differences in signaling pathways exist. With this aim in mind, we investigated by using transgenic mice the mechanisms through the simultaneous deficiency of both IL-1ß and IL-18 could be more protective compared to blocking the single cytokine IL-1ß or IL-18 during colitis. Colitis was provoked in mice by instillation of dinitrobenzene sulfonic acid (DNBS) in the colon. The results indicated that single knockout (KO) mice of IL-1ß or IL-18, and double KO mice of both IL-1ß and IL-18 were hyporesponsive to DNBS-induced colitis compared to wild type (WT) mice, in which double KO were less sensitive than single KO mice. Moreover, treatment with Anakinra (IL-1R antagonist) also ameliorated colitis, in views of macroscopic and histological alteration, infiltration of neutrophils or Th1 cells, oxidative and nitrosative stress. Anakinra more significantly reduced cyclooxygenase (COX-2) and nuclear factor (NF-κB) levels as well as IKB-α degradation compared to blocking IL-18. On the contrary, the absence of IL-18 reduced p-ERK and p-p38 mitogen-activated protein kinase (MAPKs) in a more significant way compared to blocking IL-1ß. Thus, the double KO increased the protective effects against colon inflammation maybe because different converging inflammatory pathways are being inhibited. In conclusion, the blocking of both IL-1ß and IL-18 function may be advantageous in the treatment of IBD or inflammatory diseases.


Assuntos
Colite/induzido quimicamente , Colite/metabolismo , Dinitrofluorbenzeno/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Interleucina-18/deficiência , Interleucina-1beta/deficiência , Animais , Colite/tratamento farmacológico , Dinitrofluorbenzeno/toxicidade , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Interleucina-18/antagonistas & inibidores , Interleucina-1beta/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Inflammation ; 41(2): 667-676, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29264744

RESUMO

To investigate the potential role of interleukin-18 (IL-18) in immunomodulation during tumorigenesis of esophageal carcinoma and elucidate the underlying molecular mechanism, we employed IL-18 knockout mice for this purpose. Carcinogen 4-nitroquinoline 1-oxide (4NQO) was administrated in drinking water to induce occurrence of esophageal squamous cell carcinoma (ESCC). T cell activation as indicated by the surface CD molecules was analyzed with flow cytometry. The serous content of interferon-γ (IFN-γ) along with other cytokines was determined by inflammatory human cytokine cytometric bead array. The cytotoxicity assay was performed by co-culture of tumor cells with immune cells and relative cell viability was determined by lactate dehydrogenase (LDH) assay. Apoptotic cells were stained with Annexin-V/propidium iodide (PI) and analyzed by flow cytometry. Cell proliferation was measured with Cell Counting Kit-8 (CCK-8) assay. Our data demonstrated that deficiency of IL-18 promoted the progression and development of 4NQO-induced ESCC. Loss of IL-18 suppressed the activation of T cells in the esophagus. Deficiency of IL-18 inhibited the IFN-γ production by CD8+ T cells and natural killer (NK) cells. Absence of IL-18 inhibited the cytotoxicity of CD8+ T cells and NK cell in vitro. Moreover, deficiency of IL-18 promoted the apoptosis of CD8+ T cells and inhibited the proliferation of CD8+ T cells in vitro. Our data elucidated the immunomodulatory role of IL-18 during tumorigenesis of ESCC, whose deficiency compromised antitumor immunity and contributed to immune escape of esophageal carcinoma. Our results also indicated the therapeutic potential of exogenous IL-18 against ESCC, which warrants further investigations.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Neoplasias Esofágicas/etiologia , Interferon gama/antagonistas & inibidores , Interleucina-18/deficiência , Células Matadoras Naturais/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/imunologia , Neoplasias Esofágicas/imunologia , Humanos , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Knockout
7.
J Neuroimmunol ; 314: 58-66, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29195684

RESUMO

Preclinical and clinical evidence suggests pro-inflammatory cytokines might play an important role in the neurobiology of schizophrenia and stress-related psychiatric disorders. Interleukin-18 (IL-18) is a member of the IL-1 family of cytokines and it is widely expressed in brain regions involved in emotional regulation. Since IL-18 involvement in the neurobiology of mental illnesses, including schizophrenia, remains unknown, this work aimed at investigating the behavior of IL-18 null mice (KO) in different preclinical models: 1. the prepulse inhibition test (PPI), which provides an operational measure of sensorimotor gating and schizophrenic-like phenotypes; 2. amphetamine-induced hyperlocomotion, a model predictive of antipsychotic activity; 3. resident-intruder test, a model predictive of aggressive behavior. Furthermore, the animals were submitted to models used to assess depressive- and anxiety-like behavior. IL-18KO mice showed impaired baseline PPI response, which was attenuated by d-amphetamine at a dose that did not modify PPI response in wild-type (WT) mice, suggesting a hypodopaminergic prefrontal cortex function in those mice. d-Amphetamine, however, induced hyperlocomotion in IL-18KO mice compared to their WT counterparts, suggesting hyperdopaminergic activity in the midbrain. Moreover, IL-18KO mice presented increased basal levels of IL-1ß levels in the hippocampus and TNF-α in the prefrontal cortex, suggesting an overcompensation of IL-18 absence by increased levels of other proinflammatory cytokines. Although no alteration was observed in the forced swimming or in the elevated plus maze tests in naïve IL-18KO mice, these mice presented anxiogenic-like behavior after exposure to repeated forced swimming stress. In conclusion, deletion of the IL-18 gene resembled features similar to symptoms observed in schizophrenia (positive and cognitive symptoms, aggressive behavior), in addition to increased susceptibility to stress. The IL-18KO model, therefore, could provide new insights into how changes in brain immunological homeostasis induce behavioral changes related to psychiatric disorders, such as schizophrenia.


Assuntos
Encéfalo/imunologia , Interleucina-18/deficiência , Interleucina-18/imunologia , Esquizofrenia/imunologia , Animais , Comportamento Animal/fisiologia , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Masculino , Transtornos Mentais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Esquizofrenia/genética
8.
J Neurovirol ; 23(6): 845-854, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28895072

RESUMO

The inflammasome, a cytosolic protein complex that mediates the processing and secretion of pro-inflammatory cytokines, is one of the first responders during viral infection. The cytokines secreted following inflammasome activation, which include IL-1 and IL-18, regulate cells of both the innate and adaptive immune system, guiding the subsequent immune responses. In this study, we used murine coronavirus, mouse hepatitis virus (MHV), infection of the central nervous system and liver to assess of the role of the inflammasome and its related cytokines on pathogenesis and host defense during viral infection. Mice lacking all inflammasome signaling due to the absence of caspase-1 and -11 were more vulnerable to infection, with poor survival and elevated viral replication compared to wild-type mice. Mice lacking IL-1 signaling experienced elevated viral replication but similar survival compared to wild-type controls. In the absence of IL-18, mice had elevated viral replication and poor survival, and this protective effect of IL-18 was found to be due to promotion of interferon gamma production in αß T cells. These data suggest that inflammasome signaling is largely protective during murine coronavirus infection, in large part due to the pro-inflammatory effects of IL-18.


Assuntos
Infecções por Coronavirus/imunologia , Interleucina-18/imunologia , Interleucina-1/imunologia , Vírus da Hepatite Murina/imunologia , Transdução de Sinais/imunologia , Imunidade Adaptativa , Animais , Caspase 1/deficiência , Caspase 1/genética , Caspase 1/imunologia , Caspases/deficiência , Caspases/genética , Caspases/imunologia , Caspases Iniciadoras , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Regulação da Expressão Gênica , Imunidade Inata , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interferon gama/genética , Interferon gama/imunologia , Interleucina-1/deficiência , Interleucina-1/genética , Interleucina-18/deficiência , Interleucina-18/genética , Fígado/imunologia , Fígado/patologia , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/patogenicidade , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/virologia , Carga Viral , Replicação Viral
9.
Int J Cardiol ; 243: 396-403, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28526544

RESUMO

BACKGROUND: Interleukin-18 (IL-18) neutralization protects against lipopolysaccharide (LPS)-induced injuries, including myocardial dysfunction. However, the mechanism is yet to be fully elucidated. The aim of the present study was to determine whether IL-18 gene deletion prevents sepsis-induced cardiac dysfunction and to elucidate the potential mechanisms underlying IL-18-mediated cardiotoxicity by LPS. METHODS AND RESULTS: Ten-week-old male wild-type (WT) and IL-18 knockout (IL-18 KO) mice were intraperitoneally administered LPS. Serial echocardiography showed better systolic pump function and less left ventricular (LV) dilatation in LPS-treated IL-18 KO mice compared with those in LPS-treated WT mice. LPS treatment significantly decreased the levels of phospholamban (PLN) and Akt phosphorylation in WT mice compared with those in saline-treated WT mice, while the LPS-induced decrease in the phosphorylation levels was attenuated in IL-18 KO mice compared with that in WT mice. IL-18 gene deletion also attenuated an LPS-induced increase of type 2 protein phosphatase 2A (PP2A) activity, a molecule that dephosphorylates PLN and Akt. There was no difference in type 1 protein phosphatase (PP1) activity. To address whether IL-18 affects PLN and Akt phosphorylation via PP2A activation in cardiomyocytes, rat neonatal cardiac myocytes were cultured and stimulated using 100ng/ml of recombinant rat IL-18. Exogenous IL-18 decreased the level of PLN and Akt phosphorylation in cardiomyocytes. PP2A activity but not PP1 activity was increased by IL-18 stimulation in cardiomyocytes. CONCLUSIONS: IL-18 plays a pivotal role in advancing sepsis-induced cardiac dysfunction, and the mechanisms underlying IL-18-mediated cardiotoxicity potentially involve the regulation of PLN and Akt phosphorylation through PP2A activity.


Assuntos
Deleção de Genes , Cardiopatias/metabolismo , Interleucina-18/deficiência , Interleucina-18/genética , Proteína Fosfatase 2/metabolismo , Sepse/metabolismo , Animais , Células Cultivadas , Ativação Enzimática/fisiologia , Cardiopatias/genética , Cardiopatias/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Sepse/genética , Sepse/prevenção & controle
10.
Cytokine ; 96: 132-137, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28399485

RESUMO

Previous study has demonstrated that the NLRP3 inflammasome is essential for protecting murine host against Enterovirus 71 (EV71) infection. However, the underlying mechanism remained unknown. Here we discovered that the pleiotropic cytokine interleukin-18 (IL-18), an NLRP3 inflammasome-dependent effector protein, exhibits a protective capability against EV71 challenge. Deficiency of IL-18 in mice exacerbated EV71 infection, which was reflected by increased viral replication, elevated production of interferons (IFN-ß, IFN-γ), proinflammatory cytokines (TNF-α, IL-6) and chemokine CCL2,as well as decreased survival of experimental animals. Conversely, administration of recombinant IL-18 considerably restrained EV71 infection in IL-18 deficient mice. Thus, our results revealed a protective role for IL-18 against EV71 challenge, and indicated a novel therapeutic application for IL-18 in EV71 associated hand, foot, and mouth disease (HFMD).


Assuntos
Enterovirus/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Interleucina-18/administração & dosagem , Interleucina-18/uso terapêutico , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Doença de Mão, Pé e Boca/tratamento farmacológico , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/virologia , Fatores Imunológicos , Inflamassomos , Interferons/biossíntese , Interferons/genética , Interferons/imunologia , Interleucina-18/deficiência , Interleucina-18/genética , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
11.
Shock ; 47(4): 455-462, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27648697

RESUMO

BACKGROUND: Dysregulation of glucose metabolism, including hyperglycemia with insulin resistance, is commonly observed in critically ill patients. Interleukin-18 (IL-18) improves the insulin resistance associated with obesity, but the relationship between IL-18 and glucose metabolism in sepsis was unclear. The purpose of this study was to investigate the influence of IL-18 on hyperglycemia during sepsis. METHODS: Sepsis was induced using cecal ligation and puncture (CLP) in wild-type (WT) mice, IL-18 knockout (KO) mice, and IL-18 KO mice pretreated with recombinant IL-18. Blood glucose and plasma insulin, glucagon, and corticosterone were measured. The mRNAs for gluconeogenic enzymes (g6pc, pck1) and activation of insulin signaling were also analyzed. RESULTS: In both WT and IL-18 KO mice, CLP operation led to hyperglycemia that lasted longer (18 h) than after sham operation (6 h). Blood glucose levels in IL-18 KO mice were significantly higher than in WT mice, without alteration of insulin or glucagon levels. In IL-18 KO mice, insulin signaling in the liver and skeletal muscle was decreased during hyperglycemia as compared with WT mice without suppression of hepatic glucose production enzymes. Pretreatment with recombinant IL-18 reduced blood glucose levels after CLP. Additionally, corticosterone levels were higher after CLP in the presence of either endogenous or exogenous IL-18. CONCLUSION: IL-18 may reduce blood glucose by modulating insulin signaling in the liver during sepsis-induced hyperglycemia. IL-18 is an important factor associated with alterations in blood glucose during sepsis.


Assuntos
Glicemia/efeitos dos fármacos , Corticosterona/sangue , Interleucina-18/metabolismo , Interleucina-18/uso terapêutico , Sepse/sangue , Animais , Modelos Animais de Doenças , Glucose-6-Fosfatase/genética , Insulina/sangue , Interleucina-18/deficiência , Interleucina-18/genética , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Sepse/tratamento farmacológico , Sepse/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
12.
Clin Sci (Lond) ; 130(19): 1727-39, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27413021

RESUMO

Interleukin (IL)-18 is a member of the IL-1 family of cytokines and was described originally as an interferon γ-inducing factor. Aldosterone plays a central role in the regulation of sodium and potassium homoeostasis by binding to the mineralocorticoid receptor and contributes to kidney and cardiovascular damage. Aldosterone has been reported to induce IL-18, resulting in cardiac fibrosis with induced IL-18-mediated osteopontin (OPN). We therefore hypothesized that aldosterone-induced renal fibrosis via OPN may be mediated by IL-18. To verify this hypothesis, we compared mice deficient in IL-18 and wild-type (WT) mice in a model of aldosterone/salt-induced hypertension. IL-18(-/-) and C57BL/6 WT mice were used for the uninephrectomized aldosterone/salt hypertensive model, whereas NRK-52E cells (rat kidney epithelial cells) were used in an in vitro model. In the present in vivo study, IL-18 protein expression was localized in medullary tubules in the WT mice, whereas in aldosterone-infused WT mice this expression was up-regulated markedly in the proximal tubules, especially in injured and dilated tubules. This renal damage caused by aldosterone was attenuated significantly by IL-18 knockout with down-regulation of OPN expression. In the present in vitro study, aldosterone directly induced IL-18 gene expression in renal tubular epithelial cells in a concentration- and time-dependent manner. These effects were inhibited completely by spironolactone. IL-18 may be a key mediator of aldosterone-induced renal fibrosis by inducing OPN, thereby exacerbating renal interstitial fibrosis. Inhibition of IL-18 may therefore provide a potential target for therapeutic intervention aimed at preventing the progression of renal injury.


Assuntos
Aldosterona/administração & dosagem , Interleucina-18/deficiência , Animais , Pressão Sanguínea/efeitos dos fármacos , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Fibrose/patologia , Fibrose/fisiopatologia , Humanos , Interleucina-18/genética , Rim/metabolismo , Rim/patologia , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/genética , Osteopontina/metabolismo , Potássio/administração & dosagem , Sódio/administração & dosagem , Espironolactona/administração & dosagem
13.
Transl Res ; 173: 101-114.e7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27063959

RESUMO

We investigated potential pathophysiological relationships between interleukin 18 (IL-18) and dyslipidemia, nonalcoholic fatty liver disease (NAFLD) or nonalcoholic steatohepatitis (NASH). Compared with Il18(+/+) mice, IL-18 knockout (Il18(-/-)) mice developed hypercholesterolemia and hyper-high-density-lipoprotein-cholesterolemia as well as hypertriglyceridemia as they aged, and these disorders occurred before the manifestation of obesity and might cause secondary NASH. The analyses of molecular mechanisms involved in the onset of dyslipidemia, NAFLD, and NASH in Il18(-/-) mice identified a number of genes associated with these metabolic diseases. In addition, molecules related to circadian rhythm might affect these extracted genes. The intravenous administration of recombinant IL-18 significantly improved dyslipidemia, inhibited the body weight gain of Il18(+/+) mice, and prevented the onset of NASH. The expression of genes related to these dysfunctions was also affected by recombinant IL-18 administration. In conclusion, this study demonstrated the critical function of IL-18 in lipid metabolism and these findings might contribute to the progress of novel treatments for NAFLD or NASH.


Assuntos
Dislipidemias/complicações , Fígado Gorduroso/complicações , Interleucina-18/deficiência , Hepatopatia Gordurosa não Alcoólica/complicações , Envelhecimento/patologia , Animais , Peso Corporal/efeitos dos fármacos , Ritmo Circadiano/efeitos dos fármacos , Dislipidemias/sangue , Dislipidemias/genética , Dislipidemias/patologia , Fígado Gorduroso/sangue , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Interleucina-18/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipídeos/biossíntese , Lipídeos/sangue , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/sangue , Obesidade/complicações , Obesidade/genética , Obesidade/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Recombinantes/farmacologia
15.
Cell Rep ; 13(9): 1922-36, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26655906

RESUMO

Microbial pattern molecules in the intestine play immunoregulatory roles via diverse pattern recognition receptors. However, the role of the cytosolic DNA sensor AIM2 in the maintenance of intestinal homeostasis is unknown. Here, we show that Aim2(-/-) mice are highly susceptible to dextran sodium sulfate-induced colitis that is associated with microbial dysbiosis as represented by higher colonic burden of commensal Escherichia coli. Colonization of germ-free mice with Aim2(-/-) mouse microbiota leads to higher colitis susceptibility. In-depth investigation of AIM2-mediated host defense responses reveals that caspase-1 activation and IL-1ß and IL-18 production are compromised in Aim2(-/-) mouse colons, consistent with defective inflammasome function. Moreover, IL-18 infusion reduces E. coli burden as well as colitis susceptibility in Aim2(-/-) mice. Altered microbiota in inflammasome-defective mice correlate with reduced expression of several antimicrobial peptides in intestinal epithelial cells. Together, these findings implicate DNA sensing by AIM2 as a regulatory mechanism for maintaining intestinal homeostasis.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Colo/metabolismo , Proteínas de Ligação a DNA/genética , DNA/metabolismo , Animais , Caspase 1/deficiência , Caspase 1/genética , Caspase 1/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Colo/microbiologia , Citocinas/genética , Citocinas/metabolismo , DNA/química , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Sulfato de Dextrana/toxicidade , Suscetibilidade a Doenças , Disbiose , Escherichia coli/patogenicidade , Fezes/microbiologia , Inflamassomos/metabolismo , Interleucina-18/deficiência , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/deficiência , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
16.
Sci Rep ; 5: 17977, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26656097

RESUMO

Brown and beige adipocytes recruitment in brown (BAT) or white adipose tissue, mainly in the inguinal fat pad (iWAT), meet the need for temperature adaptation in cold-exposure conditions and protect against obesity in face of hypercaloric diets. Using interleukin18 (Il18) and Il18 receptor 1- knockout (Il18r1-KO) mice, this study aimed to investigate the role of IL18 signaling in BAT and iWAT activation and thermogenesis under both stimuli. Il18-KO, extremely dietary obesity-prone as previously described, failed to develop diet-induced thermogenesis as assessed by BAT and iWAT Ucp1 mRNA levels. Overweight when fed standard chow but not HFD, HFD-fed Il18r1-KO mice exhibited increased iWAT Ucp1 gene expression. Energy expenditure was reduced in pre-obese Il18r1-KO mice and restored upon HFD-challenge. Cold exposure lead to similar results; Il18r1-KO mice were protected against acute body temperature drop, displaying a more brown-like structure, alternative macrophage activation and thermogenic gene expression in iWAT than WT controls. Opposite effects were observed in Il18-KO mice. Thus, Il18 and Il18r1 genetic ablation disparate effects on energy homeostasis are likely mediated by divergent BAT responses to thermogenic stimuli as well as iWAT browning. These results suggest that a more complex receptor-signaling system mediates the IL18 adipose-tissue specific effects in energy expenditure.


Assuntos
Tecido Adiposo Marrom/patologia , Interleucina-18/deficiência , Receptores de Interleucina-18/deficiência , Gordura Subcutânea/fisiologia , Termogênese , Tecido Adiposo Marrom/efeitos dos fármacos , Animais , Temperatura Baixa , Dieta Hiperlipídica , Metabolismo Energético , Expressão Gênica , Interleucina-18/administração & dosagem , Camundongos , Camundongos Knockout , Fenótipo , Termogênese/genética
17.
Biomed Res Int ; 2015: 318306, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25918710

RESUMO

Perinatal infections have a negative impact on brain development. However, the underlying mechanisms leading to neurological impairment are not completely understood and reliable models of inflammation are urgently needed. Using phorbol-myristate-acetate as an activator of inflammation, we investigated the effect on the developing rodent brain. Neonatal rats and mice deficient in IL-18 or IRAK-4 were exposed to PMA. Brains were assessed for regulation of pro- and anti-inflammatory cytokines and cell death 24 hrs, 7 and 14 days after treatment. PMA induced an inflammatory response and caused widespread neurodegeneration in the brains of 3- and 7-day-old rats. In contrast, 14-day-old rats were resistant to the neurotoxic effect of PMA. Histological evaluation at the age of 14 and 21 days revealed a destruction of the cortical microstructure with decreased numerical density of neuronal cells. Mice deficient in IL-18 or IRAK-4 were protected against PMA induced brain injury. PMA treatment during a vulnerable period can alter brain development. IL-18 and IRAK-4 appear to be important for the development of PMA induced injury.


Assuntos
Lesões Encefálicas/genética , Encéfalo/crescimento & desenvolvimento , Inflamação/fisiopatologia , Quinases Associadas a Receptores de Interleucina-1/genética , Interleucina-18/genética , Animais , Encéfalo/efeitos dos fármacos , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/fisiopatologia , Feminino , Inflamação/induzido quimicamente , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/deficiência , Interleucina-18/deficiência , Camundongos , Ésteres de Forbol/toxicidade , Gravidez , Ratos
18.
J Leukoc Biol ; 97(4): 665-75, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25548255

RESUMO

IL-18 is a proinflammatory and immune regulatory cytokine, member of the IL-1 family. IL-18 was initially identified as an IFN-γ-inducing factor in T and NK cells, involved in Th1 responses. IL-18 is produced as an inactive precursor (pro-IL-18) that is enzymatically processed into a mature form by Casp1. Different cells, such as macrophages, DCs, microglial cells, synovial fibroblasts, and epithelial cells, express pro-IL-18, and the production of bioactive IL-18 is mainly regulated at the processing level. PAMP or DAMP molecules activate inflammasomes, which trigger Casp1 activation and IL-18 conversion. The natural inhibitor IL-18BP , whose production is enhanced by IFN-γ and IL-27, further regulates IL-18 activity in the extracellular environment. Inflammasomes and IL-18 represent double-edged swords in cancer, as their activation may promote tumor development and progression or oppositely, enhance anti-tumor immunity and limit tumor growth. IL-18 has shown anti-tumor activity in different preclinical models of cancer immunotherapy through the activation of NK and/or T cell responses and has been tested in clinical studies in cancer patients. However, the dual role of IL-18 in different experimental tumor models and human cancers raises critical issues on its therapeutic use in cancer. This review will summarize the biology of the IL-18/IL-18R/IL-18BP system and will address the role of IL-18 and its inhibitor, IL-18BP, in cancer biology and immunotherapy.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Interleucina-18/fisiologia , Neoplasias/metabolismo , Animais , Caspase 1/fisiologia , Ensaios Clínicos Fase I como Assunto , Cocarcinogênese , Progressão da Doença , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática , Humanos , Imunoterapia , Imunoterapia Adotiva , Inflamassomos/fisiologia , Inflamação/imunologia , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Interleucina-18/deficiência , Interleucina-18/uso terapêutico , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/microbiologia , Fagócitos/imunologia , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores de Interleucina-18/fisiologia , Viroses/imunologia , Viroses/metabolismo
19.
J Invest Dermatol ; 134(10): 2562-2569, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24756108

RESUMO

IFN-γ has a central role in the defense against infections and cancer. More recently, however, IFN-γ has also been reported to have immunosuppressive effects in models of autoimmune disease, melanoma, and premalignant skin disease. Although IL-12 and IL-18 are critical inducers of IFN-γ during infection, the mechanisms that induce IFN-γ in an immunosuppressive context are unknown. Previously, we identified a key role for IFN-γ in mediating the suppression of antigen-specific immune responses in a transgenic mouse model of human papillomavirus (HPV)-associated epidermal hyperplasia, driven by the expression of the HPV16 E7 oncoprotein from a keratin 14 promoter (K14E7). We now demonstrate elevated production of IFN-γ, IL-18, and IL-12 by K14E7 transgenic skin compared with nontransgenic skin. IFN-γ in K14E7 transgenic skin was produced predominantly by CD8(+) and CD4(+) T cells, which were present in greater numbers in K14E7 transgenic skin. Production of IFN-γ in K14E7 skin required IL-18 but not IL-12. Our findings show that IL-18 contributes to inducing IFN-γ in an immunosuppressive cutaneous environment caused by viral oncogene-driven hyperplasia.


Assuntos
Papillomavirus Humano 16 , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Dermatopatias Virais/metabolismo , Pele/metabolismo , Pele/patologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Modelos Animais de Doenças , Feminino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Hiperplasia/metabolismo , Hiperplasia/patologia , Terapia de Imunossupressão , Interleucina-12/deficiência , Interleucina-12/genética , Interleucina-18/deficiência , Interleucina-18/genética , Queratina-14/genética , Queratina-14/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Dermatopatias Virais/patologia
20.
Am J Physiol Heart Circ Physiol ; 306(7): H1025-31, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24531812

RESUMO

Patients with heart failure (HF) have enhanced systemic IL-1 activity, and, in the experimental mouse model, IL-1 induces left ventricular (LV) systolic dysfunction. Whether the effects of IL-1 are direct or mediated by an inducible cytokine, such as IL-18, is unknown. Recombinant human IL-18-binding protein (IL-18BP) or an IL-18-blocking antibody (IL-18AB) was used to neutralize endogenous IL-18 after challenge with the plasma of patients with HF or with recombinant murine IL-1ß in adult male mice. Plasma levels of IL-18 and IL-6 (a key mediator of IL-1-induced systemic effects) and LV fractional shortening were measured in mice sedated with pentobarbital sodium (30-50 mg/kg). Mice with genetic deletion of IL-18 or IL-18 receptors were compared with matching wild-type mice. A group of mice received murine IL-18 to evaluate the effects on LV fractional shortening. Plasma from HF patients and IL-1ß induced LV systolic dysfunction that was prevented by pretreatment with IL-18AB or IL-18BP. IL-1ß failed to induce LV systolic dysfunction in mice with genetic deletion of IL-18 signaling. IL-1ß induced a significant increase in plasma IL-18 and IL-6 levels. Genetic or pharmacological inhibition of IL-18 signaling failed to block the induction of IL-6 by IL-1ß. In conclusion, IL-1 induces a release of active IL-18 in the mouse that mediates the LV systolic dysfunction but not the induction of IL-6. IL-18 blockade may therefore represent a novel and more targeted therapeutic approach to treat HF.


Assuntos
Insuficiência Cardíaca/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta , Disfunção Ventricular Esquerda/metabolismo , Função Ventricular Esquerda , Animais , Anticorpos/farmacologia , Modelos Animais de Doenças , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-18/deficiência , Interleucina-18/genética , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Sístole , Fatores de Tempo , Disfunção Ventricular Esquerda/sangue , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA