Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunopharmacol Immunotoxicol ; 45(4): 485-496, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36757290

RESUMO

BACKGROUND: Despite its homeostatic role, inflammation is involved in several pathologies, such as acute lung injury. Morita-Ballys-Hilman adducts (MBHA) are a group of synthetic molecules and present a wide range of biological activities, including anti-inflammatory action. Thus, this study aimed to assess whether ISACN, an MBHA, modulates inflammation during acute lung injury induced by lipopolysaccharide (LPS). METHODS: BALB/c mice were intraperitoneally treated with 24 mg/kg ISACN and challenged with LPS (2.5 mg/kg). On bronchoalveolar lavage fluid (BALF), we assessed the total and differential leukocyte count and measurement of protein leakage, cytokines (IL-1ß, IL-6, and TNF-α), and chemokine (CXCL-1). Additionally, lung histopathology was also performed (H&E staining). In vitro studies were conducted with peritoneal macrophages to assess the possible mechanism of action. They were cultured in the presence of ISACN (5 and 10 µM) and stimulated by LPS (1 µg/mL). RESULTS: ISACN reduced neutrophil migration, protein leakage, and inflammatory cytokines (IL-1ß, IL-6, and TNF-α) without interfering with the production of CXCL1. In addition, ISACN caused a decrease in LPS-induced lung injury as evident from histopathological changes. In peritoneal macrophages, ISACN diminishes the nitric oxide and cytokine levels (IL-1ß, IL-6, and TNF-α). The treatment with ISACN (10 µM) also reduced LPS-induced TLR4, CD69, iNOS overexpression, and the LPS-induced ERK, JNK, and p38 phosphorylation. CONCLUSION: Thus, this work showed for the first time the immunomodulatory action of MBHA in LPS-induced acute lung injury and provided new evidence for the mechanisms related to the anti-inflammatory effect of ISACN.


Assuntos
Acrilonitrila , Lesão Pulmonar Aguda , Camundongos , Animais , Lipopolissacarídeos/toxicidade , Acrilonitrila/efeitos adversos , Acrilonitrila/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Pulmão , Citocinas/metabolismo , Anti-Inflamatórios/uso terapêutico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo
2.
Nat Prod Res ; 36(22): 5879-5883, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34963384

RESUMO

Patulin (PAT) is a natural product isolated from several species of fungi. Here, we evaluated the effect of PAT (62.5-4,000 ng/ml) in lipopolysaccharide (LPS)-activated murine peritoneal macrophages. Cell viability assay showed that PAT at concentrations up to 250 ng/ml did not affect macrophage viability. PAT (250 ng/ml) significantly reduced LPS-induced nitric oxide production (by 98.4%), inducible nitric oxide synthase (iNOS) expression (by 83.5%), and iNOS messenger ribonucleic acid expression (by 100.0%). Moreover, PAT significantly reduced LPS-induced interleukin-1ß (by 80.6%), cluster of differentiation (CD) 69 (by 63.1%), and Toll-like receptor (TLR) 4 (by 91.9%) protein expression. Finally, PAT significantly reduced LPS-triggered phosphorylation of all mitogen-activated protein kinases (MAPK) assessed: extracellular signal-regulated kinase (ERK; by 89.5%), c-Jun N-terminal kinase (JNK; by 77.5%), and p38 (by 72.3%). Taken together, these data suggest that PAT downregulates acute inflammatory response, inhibiting nitric oxide production by suppressing CD69-TLR4/ERK-JNK-p38 MAPKs/Nos2/iNOS signaling pathway.


Assuntos
Lipopolissacarídeos , Patulina , Animais , Camundongos , Lipopolissacarídeos/farmacologia , Óxido Nítrico , Patulina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais , NF-kappa B/metabolismo
3.
Int Immunopharmacol ; 90: 107233, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33290963

RESUMO

The SARS-Cov2 infection triggers a multisystem inflammatory disorder, knowing as COVID-19, a pandemic disease. This disease is characterized by acute respiratory distress syndrome, cytokine-driven hyperinflammation, and leukocytes count changes. The innate immune response has been linked to COVID-19 immunopathogenesis (e.g., dysfunctional IFN response and myeloid inflammation). In this regard, neutrophils have been highlighted as essential effector cells in the development of COVID-19. This review summarized the significant finds about neutrophils and its effector mechanisms (e.g., neutrophils enzymes and cytokines, neutrophil extracellular traps) in COVID-19 so far.


Assuntos
COVID-19/imunologia , Neutrófilos/imunologia , SARS-CoV-2 , Humanos , Inflamação/imunologia
4.
Int Immunopharmacol ; 83: 106399, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32193104

RESUMO

LQB 118, a hydride molecule, has been described as an antineoplastic and antiparasitic drug. Recently, LQB118 was also shown to display anti-inflammatory properties using an LPS-induced lung inflammation model. However, LQB 118 effects on the inflammatory response induced by zymosan has not been demonstrated. In this study, swiss mice were LQB 118 intraperitoneally (i.p.) treated and zymosan was used to induce peritoneal inflammation. Peritoneal fluid was collected and used for cell counting and proinflammatory cytokines quantification (IL-1ß, IL-6, and TNF-α) by immunoenzymatic assay (ELISA). For in vitro studies, peritoneal macrophages zymosan-stimulated were used. Results demonstrated that LQB 118 treatment reduced polymorphonuclear cell migration and TNF-α, IL-1ß, and IL-6 levels in the peritoneal cavity. In macrophages, LQB 118 treatment display no cytotoxic effect and is also able to reduce cytokines levels. To investigate LQB 118 putative mechanism of action, TLR2, CD69, and P-p38 MAPK expression were evaluated. LQB 118 treatment reduced CD69 expression and p38 phosphorylation induced by zymosan. Furthermore, LQB 118 was able to negatively modulate TLR2 expression in the presence of inflammatory stimulus. Thus, our study provide new evidences for the mechanisms related to the anti-inflammatory effect of LQB 118 in vivo and in vitro.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antineoplásicos/uso terapêutico , Inflamação/tratamento farmacológico , Macrófagos/imunologia , Naftoquinonas/uso terapêutico , Peritônio/imunologia , Peritonite/tratamento farmacológico , Pterocarpanos/uso terapêutico , Animais , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Zimosan/imunologia
5.
Inflammopharmacology ; 28(3): 787-793, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31087249

RESUMO

Ouabain, a hormone that inhibits Na+/K+-ATPase, modulates many aspects of the inflammatory response. It has been previously demonstrated that ouabain inhibits neutrophil migration in several inflammation models in vivo, but little is known about the mechanisms underlying this effect. Thus, this work aimed to evaluate the effect of ouabain on molecules related to neutrophil migration. For this purpose, neutrophils obtained from mouse bone marrow were treated with ouabain (1, 10, and 100 nM) in vitro. Neutrophil viability was assessed by annexin V/propidium iodide staining. Ouabain treatment did not affect neutrophil viability at different times (2, 4, and 24 h). However, basal neutrophil viability was decreased after 4 h. Thus, we assessed the effect of ouabain on the adhesion molecule CD18, an integrin ß2 chain protein, and on the chemokine receptor CXCR2 after 2 h of treatment. CD18 expression was reduced (by 30%) by 1 nM ouabain. However, the expression of CXCR2 on the neutrophil membrane was not affected by ouabain treatment (1, 10, and 100 nM). Moreover, ouabain (1, 10, and 100 nM) did not modulate the zymosan-induced secretion of CXCL1 (a chemokine receptor CXCR2 ligand) in macrophage cultures. These data suggest that the inhibitory effect of ouabain on neutrophil migration is related to reduced CD18 expression, indicating a novel mechanism of action.


Assuntos
Antígenos CD18/metabolismo , Neutrófilos/efeitos dos fármacos , Ouabaína/farmacologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL1/metabolismo , Feminino , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Neutrófilos/metabolismo
7.
Front Physiol ; 8: 895, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29176951

RESUMO

Since the discovery of ouabain as a cardiotonic steroid hormone present in higher mammals, research about it has progressed rapidly and several of its physiological and pharmacological effects have been described. Ouabain can behave as a stress hormone and adrenal cortex is its main source. Direct effects of ouabain are originated due to the binding to its receptor, the Na+/K+-ATPase, on target cells. This interaction can promote Na+ transport blockade or even activation of signaling transduction pathways (e.g., EGFR/Src-Ras-ERK pathway activation), independent of ion transport. Besides the well-known effect of ouabain on the cardiovascular system and blood pressure control, compelling evidence indicates that ouabain regulates a number of immune functions. Inflammation is a tightly coordinated immunological function that is also affected by ouabain. Indeed, this hormone can modulate many inflammatory events such as cell migration, vascular permeability, and cytokine production. Moreover, ouabain also interferes on neuroinflammation. However, it is not clear how ouabain controls these events. In this brief review, we summarize the updates of ouabain effect on several aspects of peripheral and central inflammation, bringing new insights into ouabain functions on the immune system.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA