Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38970705

RESUMO

Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRß, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRß as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.


Assuntos
Homeostase , Receptores X do Fígado , Macrófagos Alveolares , Pneumonia , Surfactantes Pulmonares , Transdução de Sinais , Animais , Receptores X do Fígado/metabolismo , Receptores X do Fígado/genética , Surfactantes Pulmonares/metabolismo , Camundongos , Pneumonia/metabolismo , Pneumonia/patologia , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pulmão/metabolismo , Pulmão/patologia , Células Epiteliais Alveolares/metabolismo , Asma/metabolismo , Asma/patologia , Asma/genética , Colesterol/metabolismo , Metabolismo dos Lipídeos , Fagocitose
2.
Front Endocrinol (Lausanne) ; 14: 1266150, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38144555

RESUMO

Preclinical and clinical studies suggest that hypothyroidism might cause hepatic endocrine and metabolic disturbances with features that mimic deficiencies of testosterone and/or GH. The absence of physiological interactions between testosterone and GH can be linked to male differentiated liver diseases. Testosterone plays relevant physiological effects on somatotropic-liver axis and liver composition and the liver is a primary organ of interactions between testosterone and GH. However, testosterone exerts many effects on liver through complex and poorly understood mechanisms. Testosterone impacts liver functions by binding to the Androgen Receptor, and, indirectly, through its conversion to estradiol, and cooperation with GH. However, the role of testosterone, and its interaction with GH, in the hypothyroid liver, remains unclear. In the present work, the effects of testosterone, and how they impact on GH-regulated whole transcriptome and lipid composition in the liver, were studied in the context of adult hypothyroid-orchiectomized rats. Testosterone replacement positively modulated somatotropic-liver axis and impacted liver transcriptome involved in lipid and glucose metabolism. In addition, testosterone enhanced the effects of GH on the transcriptome linked to lipid biosynthesis, oxidation-reduction, and metabolism of unsaturated and long-chain fatty acids (FA). However, testosterone decreased the hepatic content of cholesterol esters and triacylglycerols and increased fatty acids whereas GH increased neutral lipids and decreased polar lipids. Biological network analysis of the effects of testosterone on GH-regulated transcriptome confirmed a close connection with crucial proteins involved in steroid and fatty acid metabolism. Taken together, this comprehensive analysis of gene expression and lipid profiling in hypothyroid male liver reveals a functional interplay between testosterone and pulsed GH administration.


Assuntos
Hormônio do Crescimento , Hipotireoidismo , Animais , Masculino , Ratos , Ácidos Graxos/metabolismo , Perfilação da Expressão Gênica , Hormônio do Crescimento/metabolismo , Hipotireoidismo/complicações , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Fígado/metabolismo , Testosterona/metabolismo , Transcriptoma
3.
Biomed Pharmacother ; 157: 114060, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36455458

RESUMO

Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal cancers worldwide, but the precise intracellular mechanisms underlying the progression of this inflammation associated cancer are not well established. SOCS2 protein plays an important role in the carcinogenesis of different tumors by regulating cytokine signalling through the JAK/STAT axis. However, its role in HCC is unclear. Here, we investigate the role of SOCS2 in HCC progression and its potential as HCC biomarker. The effects of SOCS2 in HCC progression were evaluated in an experimental model of diethylnitrosamine (DEN)-induced HCC in C57BL/6 and SOCS2 deficient mice, in cultured hepatic cells, and in liver samples from HCC patients. Mice lacking SOCS2 showed higher liver tumor burden with increased malignancy grade, inflammation, fibrosis, and proliferation than their controls. Protein and gene expression analysis reported higher pSTAT5 and pSTAT3 activation, upregulation of different proteins involved in survival and proliferation, and increased levels of proinflammatory and pro-tumoral mediators in the absence of SOCS2. Clinically relevant, downregulated expression of SOCS2 was found in neoplasia from HCC patients compared to healthy liver tissue, correlating with the malignancy grade. In summary, our data show that lack of SOCS2 increases susceptibility to chemical-induced HCC and suggest the tumor suppressor role of this protein by regulating the oncogenic and inflammatory responses mediated by STAT5 and STAT3 in the liver. Hence, SOCS2 emerges as an attractive target molecule and potential biomarker to deepen in the study of HCC treatment.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Camundongos , Animais , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Camundongos Endogâmicos C57BL , Proliferação de Células , Dietilnitrosamina/toxicidade , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
4.
Atherosclerosis ; 363: 57-68, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36459823

RESUMO

BACKGROUND AND AIMS: Atherosclerosis is widely accepted to be an inflammatory disease driven by lipid accumulation and leukocyte recruitment. More recently, galectins, a family of ß-galactoside binding proteins, have been shown to play a role in leukocyte recruitment among other immunomodulatory functions. Galectin (Gal) -9, a tandem repeat type galectin expressed by the endothelium in inflammatory environments, has been proposed to promote leukocyte recruitment. However, the role of Gal-9 in the context of monocyte recruitment remains elusive. METHODS AND RESULTS: Here, we characterise the immunomodulatory role of Gal-9 in context of atherosclerosis. We show that ApoE-/-Gal-9-/- mice have a significantly reduced aortic plaque burden compared to their ApoE-/- littermate controls after 12 weeks of high fat diet. RNA sequencing data from two independent studies reveal Lgals9 expression in leukocyte clusters isolated from murine atherosclerotic plaques. Additionally, soluble Gal-9 protein induces monocyte activation and a pro-inflammatory phenotype in macrophages. Furthermore, we show that immobilised recombinant Gal-9 acts as capture and adhesion molecule for CD14+ monocytes in a ß2-integrin and glycan dependent manner, while adhesion of monocytes to stimulated endothelium is reduced when Gal-9 is knocked down. Gal-9 also facilitates enhanced recruitment of leukocytes from peripheral arterial disease (PAD) patients compared to healthy young and aged controls. We further characterise the endothelium as source of circulating Gal-9, which is increased in plasma of PAD patients compared to healthy controls. CONCLUSIONS: These results highlight a pathological role for Gal-9 as promoter of monocyte recruitment and atherosclerotic plaque progression, making it a novel target in the prevention of plaque formation and progression.


Assuntos
Aterosclerose , Placa Aterosclerótica , Camundongos , Animais , Camundongos Endogâmicos C57BL , Células Cultivadas , Aterosclerose/patologia , Placa Aterosclerótica/metabolismo , Monócitos/metabolismo
5.
Cancers (Basel) ; 14(21)2022 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-36358593

RESUMO

Tamoxifen improves the overall survival rate in hormone receptor-positive breast cancer patients. However, despite the fact that it exerts antagonistic effects on the ERα, it can act as a partial agonist, resulting in tumor growth in estrogen-sensitive tissues. In this study, highly functionalized 5-hydroxy-2H-pyrrol-2-ones were synthesized and evaluated by using ERα- and phenotype-based screening assays. Compounds 32 and 35 inhibited 17ß-estradiol (E2)-stimulated ERα-mediated transcription of the luciferase reporter gene in breast cancer cells without inhibition of the transcriptional activity mediated by androgen or glucocorticoid receptors. Compound 32 regulated E2-stimulated ERα-mediated transcription by partial antagonism, whereas compound 35 caused rapid and non-competitive inhibition. Monitoring of 2D and 3D cell growth confirmed potent antitumoral effects of both compounds on ER-positive breast cancer cells. Furthermore, compounds 32 and 35 caused apoptosis and blocked the cell cycle of ER-positive breast cancer cells in the sub-G1 and G0/G1 phases. Interestingly, compound 35 suppressed the functional activity of ERα in the uterus, as demonstrated by the inhibition of E2-stimulated transcription of estrogen and progesterone receptors and alkaline phosphatase enzymatic activity. Compound 35 showed a relatively low binding affinity with ERα. However, its antiestrogenic effect was associated with an increased polyubiquitination and a reduced protein expression of ERα. Clinically relevant, a possible combinatory therapy with compound 35 may enhance the antitumoral efficacy of 4-hydroxy-tamoxifen in ER-positive breast cancer cells. In silico ADME predictions indicated that these compounds exhibit good drug-likeness, which, together with their potential antitumoral effects and their lack of estrogenic activity, offers a pharmacological opportunity to deepen the study of ER-positive breast cancer treatment.

6.
Cardiovasc Diabetol ; 21(1): 253, 2022 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-36403025

RESUMO

Galectins are ß-galactoside-binding proteins that bind and crosslink molecules via their sugar moieties, forming signaling and adhesion networks involved in cellular communication, differentiation, migration, and survival. Galectins are expressed ubiquitously across immune cells, and their function varies with their tissue-specific and subcellular location. Particularly galectin-1, -3, and -9 are highly expressed by inflammatory cells and are involved in the modulation of several innate and adaptive immune responses. Modulation in the expression of these proteins accompany major processes in cardiovascular diseases and metabolic disorders, such as atherosclerosis, thrombosis, obesity, and diabetes, making them attractive therapeutic targets. In this review we consider the broad cellular activities ascribed to galectin-1, -3, and -9, highlighting those linked to the progression of different inflammatory driven pathologies in the context of cardiovascular and metabolic disease, to better understand their mechanism of action and provide new insights into the design of novel therapeutic strategies.


Assuntos
Aterosclerose , Doenças Metabólicas , Humanos , Galectina 1/metabolismo , Galectinas/química , Galectinas/metabolismo , Imunidade , Aterosclerose/tratamento farmacológico , Doenças Metabólicas/tratamento farmacológico
7.
Biomed Pharmacother ; 144: 112330, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34673425

RESUMO

Chronic myelogenous leukemia (CML) is a hematological malignancy that highly depends on the BCR-ABL1/STAT5 signaling pathway for cell survival. First-line treatments for CML consist of tyrosine kinase inhibitors that efficiently target BCR-ABL1 activity. However, drug resistance and intolerance are still therapeutic limitations in Ph+ cells. Therefore, the development of new anti-CML drugs that exhibit alternative mechanisms to overcome these limitations is a desirable goal. In this work, the antitumoral activity of JKST6, a naphthoquinone-pyrone hybrid, was assessed in imatinib-sensitive and imatinib-resistant human CML cells. Live-cell imaging analysis revealed JKST6 potent antiproliferative activity in 2D and 3D CML cultures. JKST6 provoked cell increase in the subG1 phase along with a reduction in the G0/G1 phase and altered the expression of key proteins involved in the control of mitosis and DNA damage. Rapid increases in Annexin V staining and activation/cleavage of caspases 8, 9 and 3 were observed after JKST6 treatment in CML cells. Of interest, JKST6 inhibited BCR-ABL1/STAT5 signaling through oncokinase downregulation that was preceded by rapid polyubiquitination. In addition, JKST6 caused a transient increase in JNK and AKT phosphorylation, whereas the phosphorylation of P38-MAPK and Src was reduced. Combinatory treatment unveiled synergistic effects between imatinib and JKST6. Notably, JKST6 maintained its antitumor efficacy in BCR-ABL1-T315I-positive cells and CML cells that overexpress BCR-ABL and even restored imatinib efficacy after a short exposure time. These findings, together with the observed low toxicity of JKST6, reveal a novel multikinase modulator that might overcome the limitations of BCR-ABL1 inhibitors in CML therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Mesilato de Imatinib/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Naftoquinonas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT5/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Fator de Transcrição STAT5/genética , Transdução de Sinais
8.
Front Oncol ; 11: 626971, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718197

RESUMO

A hallmark of cancer cells includes a metabolic reprograming that provides energy, the essential building blocks, and signaling required to maintain survival, rapid growth, metastasis, and drug resistance of many cancers. The influence of tumor microenviroment on cancer cells also results an essential driving force for cancer progression and drug resistance. Lipid-related enzymes, lipid-derived metabolites and/or signaling pathways linked to critical regulators of lipid metabolism can influence gene expression and chromatin remodeling, cellular differentiation, stress response pathways, or tumor microenviroment, and, collectively, drive tumor development. Reprograming of lipid metabolism includes a deregulated activity of mevalonate (MVA)/cholesterol biosynthetic pathway in specific cancer cells which, in comparison with normal cell counterparts, are dependent of the continuous availability of MVA/cholesterol-derived metabolites (i.e., sterols and non-sterol intermediates) for tumor development. Accordingly, there are increasing amount of data, from preclinical and epidemiological studies, that support an inverse association between the use of statins, potent inhibitors of MVA biosynthetic pathway, and mortality rate in specific cancers (e.g., colon, prostate, liver, breast, hematological malignances). In contrast, despite the tolerance and therapeutic efficacy shown by statins in cardiovascular disease, cancer treatment demands the use of relatively high doses of single statins for a prolonged period, thereby limiting this therapeutic strategy due to adverse effects. Clinically relevant, synergistic effects of tolerable doses of statins with conventional chemotherapy might enhance efficacy with lower doses of each drug and, probably, reduce adverse effects and resistance. In spite of that, clinical trials to identify combinatory therapies that improve therapeutic window are still a challenge. In the present review, we revisit molecular evidences showing that deregulated activity of MVA biosynthetic pathway has an essential role in oncogenesis and drug resistance, and the potential use of MVA pathway inhibitors to improve therapeutic window in cancer.

9.
Biomed Pharmacother ; 130: 110595, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32771893

RESUMO

Macrophages are key cells in both acute and chronic inflammatory settings. Their activation and function highly depends on the cytokines, chemokines and adhesion molecules that direct monocytes to infiltrate tissues, differentiate into macrophages, and finally lead to the clearance of such inflammatory signals. Galectins, ß-galactoside-binding lectins, are differentially expressed by various immune cells, and some members of this family have been identified as regulators of leukocyte recruitment and activation. Galectin-1 (Gal-1) and galectin-9 (Gal-9) expression has been described in immune cells, but the specific molecular mechanisms by which they modulate the inflammatory response in macrophages/monocytes are not completely understood. In this study we sought to comprehensively characterise the expression profile of endogenous Gal-1 and Gal-9 in different murine and human monocyte/macrophage populations in response to different inflammatory stimuli. All subsets of murine and human macrophages expressed significant levels of Gal-1 and -9. Interestingly, murine bone marrow derived macrophages stimulated with M2 (pro-resolution) polarising agents preferentially upregulated Gal-1, while Gal-9 expression was upregulated by M1/pro-inflammatory stimulation. However, we observed differing results in human monocyte derived macrophages. Collectively, our findings report a differential expression pattern of endogenous Gal-1 and -9 in macrophage and monocyte subsets in response to a range of inflammatory stimuli. Future studies will endeavour to elucidate whether the galectins make attractive therapeutic targets or agents for regulating the inflammatory response.


Assuntos
Galectina 1/biossíntese , Galectinas/biossíntese , Inflamação/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Adulto , Idoso , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Feminino , Humanos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
10.
ACS Chem Biol ; 14(9): 2055-2064, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31465201

RESUMO

GPR84 is an orphan G-protein-coupled receptor that is expressed on immune cells and implicated in several inflammatory diseases. The validation of GPR84 as a therapeutic target is hindered by the narrow range of available chemical tools and consequent poor understanding of GPR84 pathophysiology. Here we describe the discovery and characterization of DL-175, a potent, selective, and structurally novel GPR84 agonist and the first to display significantly biased signaling across GPR84-overexpressing cells, primary murine macrophages, and human U937 cells. By comparing DL-175 with reported GPR84 ligands, we show for the first time that biased GPR84 agonists have markedly different abilities to induce chemotaxis in human myeloid cells, while causing similar levels of phagocytosis enhancement. This work demonstrates that biased agonism at GPR84 enables the selective activation of functional responses in immune cells and delivers a high-quality chemical probe for further investigation.


Assuntos
Fatores Quimiotáticos/farmacologia , Óxidos N-Cíclicos/farmacologia , Macrófagos/efeitos dos fármacos , Piridinas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Animais , Células CHO , Linhagem Celular Tumoral , Fatores Quimiotáticos/química , Cricetulus , Óxidos N-Cíclicos/química , Humanos , Camundongos , Estrutura Molecular , Fagocitose/efeitos dos fármacos , Piridinas/química , Relação Quantitativa Estrutura-Atividade , Transdução de Sinais/efeitos dos fármacos
11.
Oncogene ; 38(24): 4657-4668, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30783189

RESUMO

The signal transducer and activator of transcription (STAT) are transcription factors that work via JAK/STAT pathway regulating the expression of genes involved in cell survival, proliferation, differentiation, development, immune response, and, among other essential biological functions, hematopoiesis. JAK/STAT signaling is strictly regulated under normal physiological conditions. However, a large group of diverse diseases has been associated to an aberrant regulation of STAT factors. Erroneous modulation of the pathway leads to constitutive STAT activation, thereby driving proliferation, inflammation, and an uncontrolled immune response. Deregulated STAT5 activation has been found in the development of many hematopoietic tumors, including chronic and acute leukemias, polycythemia vera, and lymphoma. Mutations in the kinases that phosphorylate STAT5, and/or overexpression of the upstream receptor-associated tyrosine kinases have been suggested as the main drivers of constitutive STAT5 activation. Hyper-activated STAT5 leads to the aberrant expression of its target genes including antiapoptotic, proliferative, and pro-inflammatory genes, favouring tumorigenesis. In this review, we intent to discuss the biology of JAK/STAT pathway, with particular focus on STAT5 and its crucial role in the development and progression of hematologic malignancies. Furthermore, we provide a synopsis of potential therapeutic strategies based on STAT5 activity inhibition that may represent an excellent opportunity for drug development in oncohematology.


Assuntos
Antineoplásicos/uso terapêutico , Desenvolvimento de Medicamentos , Neoplasias Hematológicas/tratamento farmacológico , Oncologia , Fator de Transcrição STAT5/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Antineoplásicos/química , Desenvolvimento de Medicamentos/tendências , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Humanos , Janus Quinases/fisiologia , Oncologia/métodos , Oncologia/tendências , Fatores de Transcrição STAT/fisiologia , Transdução de Sinais
12.
Front Pharmacol ; 9: 819, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30108504

RESUMO

Interactive relationships between metabolism, inflammation, oxidative stress, and autophagy in the vascular system play a key role in the pathogenesis of diabetic cardiovascular disease. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a stress-sensitive guarantor of cellular homeostasis, which cytoprotective contributions extend beyond the antioxidant defense. We investigated the beneficial effects and underlying mechanisms of the Nrf2 inducer tert-butyl hydroquinone (tBHQ) on diabetes-driven atherosclerosis. In the experimental model of streptozotocin-induced diabetes in apolipoprotein E-deficient mice, treatment with tBHQ increased Nrf2 activity in macrophages and vascular smooth muscle cells within atherosclerotic lesions. Moreover, tBHQ significantly decreased the size, extension and lipid content of atheroma plaques, and attenuated inflammation by reducing lesional macrophages (total number and M1/M2 phenotype balance), foam cell size and chemokine expression. Atheroprotection was accompanied by both systemic and local antioxidant effects, characterized by lower levels of superoxide anion and oxidative DNA marker 8-hydroxy-2'-deoxyguanosine, reduced expression of NADPH oxidase subunits, and increased antioxidant capacity. Interestingly, tBHQ treatment upregulated the gene and protein expression of autophagy-related molecules and also enhanced autophagic flux in diabetic mouse aorta. In vitro, Nrf2 activation by tBHQ suppressed cytokine-induced expression of pro-inflammatory and oxidative stress genes, altered macrophage phenotypes, and promoted autophagic activity. Our results reinforce pharmacological Nrf2 activation as a promising atheroprotective approach in diabetes, according to the plethora of cytoprotective mechanisms involved in the resolution of inflammation and oxidative stress, and restoring autophagy.

13.
Front Immunol ; 9: 1419, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29973940

RESUMO

GPR84 is a member of the metabolic G protein-coupled receptor family, and its expression has been described predominantly in immune cells. GPR84 activation is involved in the inflammatory response, but the mechanisms by which it modulates inflammation have been incompletely described. In this study, we investigated GPR84 expression, activation, and function in macrophages to establish the role of the receptor during the inflammatory response. We observed that GPR84 expression in murine tissues is increased by endotoxemia, hyperglycemia, and hypercholesterolemia. Ex vivo studies revealed that GPR84 mRNA expression is increased by LPS and other pro-inflammatory molecules in different murine and human macrophage populations. Likewise, high glucose concentrations and the presence of oxidized LDL increased GPR84 expression in macrophages. Activation of the GPR84 receptor with a selective agonist, 6-(octylamino) pyrimidine-2,4(1H,3H)-dione (6-n-octylaminouracil, 6-OAU), enhanced the expression of phosphorylated Akt, p-ERK, and p65 nuclear translocation under inflammatory conditions and elevated the expression levels of the inflammatory mediators TNFα, IL-6, IL-12B, CCL2, CCL5, and CXCL1. In addition, GPR84 activation triggered increased bacterial adhesion and phagocytosis in macrophages. The enhanced inflammatory response mediated by 6-OAU was not observed in GPR84-/- cells nor in macrophages treated with a selective GPR84 antagonist. Collectively, our results reveal that GPR84 functions as an enhancer of inflammatory signaling in macrophages once inflammation is established. Therefore, molecules that antagonize the GPR84 receptor may be potential therapeutic tools in inflammatory and metabolic diseases.

14.
Methods Mol Biol ; 1784: 197-214, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29761401

RESUMO

The timely recruitment of innate and adaptive immune cells to sites of inflammation and repair is essential for host defense against pathogens and repair of damaged tissues. The development of bioassays such as in vitro chemotaxis assays played an important role in the original purification of chemoattractant cytokines including interleukin-1 and the CC and CXC chemokines. The earliest chemotaxis methods were based on the principle of the Boyden chamber, first described in 1962. In this chapter we give detailed protocols for more recent techniques that allow determination of macrophage chemotaxis in real time. These techniques have given new insights into the regulation of macrophage responses to chemotaxis in vitro and in vivo.


Assuntos
Bioensaio/métodos , Movimento Celular/genética , Quimiotaxia/genética , Macrófagos/imunologia , Imunidade Adaptativa/genética , Movimento Celular/imunologia , Quimiocinas CC/genética , Quimiocinas CXC/genética , Fatores Quimiotáticos/genética , Quimiotaxia/imunologia , Humanos , Imunidade Inata/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-1/genética , Macrófagos/metabolismo
15.
Lab Invest ; 98(10): 1276-1290, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29540859

RESUMO

Oxidative stress resulting from excessive production of reactive oxygen species (ROS) or impaired antioxidant defenses is closely related to the development of diabetic vascular complications, including nephropathy and atherosclerosis. Chronic activation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathway contributes to diabetic complications by inducing expression of genes involved in cell proliferation, fibrosis, inflammation, and oxidative stress. Suppressors of cytokine signaling (SOCS) family of endogenous JAK/STAT regulators is an attractive target for therapeutic intervention. We investigated the beneficial effect of two different SOCS1-targeted therapies (adenovirus-mediated gene transfer and kinase-inhibitory region peptidomimetic) to combat oxidative stress injury in an experimental diabetes model of concomitant renal and macrovascular disease (streptozotocin-induced diabetic apolipoprotein E-deficient mouse). Diabetes resulted in progressive alteration of redox balance in mice, as demonstrated by increased ROS levels and decreased antioxidant activity, which ultimately led to renal dysfunction and vascular injury. The molecular and pathological alterations in early diabetes were partially reversed by preventive intervention with SOCS1-targeted therapies. Importantly, SOCS1 peptidomimetic provided reno- and atheroprotection in diabetic mice even in a setting of established disease. Compared with untreated controls, kidney and aorta from SOCS1-treated mice exhibited significantly lower levels of superoxide anion, DNA oxidation marker and NADPH oxidase (Nox) subunits, along with higher expression of antioxidant enzymes. These trends correlated with a reduction in parameters of renal damage (albuminuria, creatinine and tubular injury), atherosclerosis (lesion size) and inflammation (leukocytes and chemokines). Mechanistic studies in renal, vascular and phagocytic cells exposed to cytokines and high-glucose showed that SOCS1 blocked ROS generation by inhibiting both Nox complex assembly and Nox subunit expression, an effect mediated by inactivation of JAK2, STAT1, and PI3K signaling pathways. This study provides evidence for SOCS1-targeted therapies, especially SOCS1 peptidomimetic, as an alternative antioxidant strategy to limit the progression of diabetic micro- and macrovascular complications.


Assuntos
Angiopatias Diabéticas/terapia , Nefropatias Diabéticas/terapia , Estresse Oxidativo , Proteína 1 Supressora da Sinalização de Citocina/fisiologia , Animais , Antioxidantes/metabolismo , Aorta/metabolismo , Terapia Genética , Rim/metabolismo , Masculino , Camundongos , NADPH Oxidases/metabolismo , Peptidomiméticos/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/metabolismo
16.
Antioxid Redox Signal ; 29(3): 237-256, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29117706

RESUMO

SIGNIFICANCE: Great attention has been placed on the link between metabolism and immune function giving rise to the term "immunometabolism." It is widely accepted that inflammation and oxidative stress are key processes that underlie metabolic complications during obesity, diabetes, and atherosclerosis. Therefore, identifying the mechanisms and mediators that are involved in the regulation of both inflammation and metabolic homeostasis is of high scientific and therapeutic interest. Recent Advances: G protein-coupled receptors (GPCRs) that signal in response to metabolites have emerged as attractive therapeutic targets in inflammatory disease. Critical Issues and Future Directions: In this review, we discuss recent findings about the physiological role of the main metabolite-sensing GPCRs, their implication in immunometabolic disorders, their principal endogenous and synthetic ligands, and their potential as drug targets in inflammation and metabolic disease. Antioxid. Redox Signal. 29, 237-256.


Assuntos
Inflamação/metabolismo , Doenças Metabólicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos
17.
Front Immunol ; 8: 1621, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29209334

RESUMO

Chemerin is a chemotactic protein that induces migration of several immune cells including macrophages, immature dendritic cells, and NK cells. Chemerin binds to three G protein-coupled receptors (GPCRs), including CCRL2. The exact function of CCRL2 remains unclear. CCRL2 expression is rapidly upregulated during inflammation, but it lacks the intracellular DRYLAIV motif required for classical GPCR downstream signalling pathways, and it has not been reported to internalise chemerin upon binding. The aim of this study was to investigate what role if any CCRL2 plays during acute inflammation. Using the zymosan- and thioglycollate-induced murine models of acute inflammation, we report that mice deficient in the Ccrl2 gene display exaggerated local and systemic inflammatory responses, characterised by increased myeloid cell recruitment. This amplified myeloid cell recruitment was associated with increased chemerin and CXCL1 levels. Furthermore, we report that the inflammatory phenotype observed in these mice is dependent upon elevated levels of endogenous chemerin. Antibody neutralisation of chemerin activity in Ccrl2-/- mice abrogated the amplified inflammatory responses. Importantly, chemerin did not directly recruit myeloid cells but rather increased the production of other chemotactic proteins such as CXCL1. Administration of recombinant chemerin to wild-type mice before inflammatory challenge recapitulated the increased myeloid cell recruitment and inflammatory mediator production observed in Ccrl2-/- mice. We have demonstrated that the absence of CCRL2 results in increased levels of local and systemic chemerin levels and exacerbated inflammatory responses during acute inflammatory challenge. These results further highlight the importance of chemerin as a therapeutic target in inflammatory diseases.

18.
Mediators Inflamm ; 2017: 4315412, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28852269

RESUMO

The endocannabinoid system consists of endogenous lipid mediators and cannabinoid receptors (CB) 1 and 2. It has previously been demonstrated that activation of the leukocyte-expressed CB2 has anti-inflammatory effects in vivo. Here, we report its role under baseline conditions and in a model of low-dose endotoxemia by comparing CB2 knockout to littermate control mice. CB2-deficient mice displayed significantly more neutrophils and fewer monocytes in the bone marrow under steady state. In initial validation experiments, administration of 1 mg/kg LPS to male C57BL/6J mice was shown to transiently upregulate systemic proinflammatory mediators (peaked at 2 hours) and mobilise bone marrow neutrophils and monocytes into circulation. In CB2 knockout mice, the level of the metalloproteinase MMP-9 was significantly elevated by 2 hours and we also observed augmented recruitment of neutrophils to the spleen in addition to increased levels of Ccl2, Ccl3, Cxcl10, and Il6. Collectively, our data show that the absence of CB2 receptor increases the levels of innate immune cell populations in the bone marrow under steady state. Furthermore, during an acute systemic inflammatory insult, we observe a highly reproducible and site-specific increase in neutrophil recruitment and proinflammatory chemokine expression in the spleen of CB2 knockout mice.


Assuntos
Endotoxemia/metabolismo , Infiltração de Neutrófilos/fisiologia , Neutrófilos/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Animais , Quimiocinas/metabolismo , Modelos Animais de Doenças , Endotoxemia/genética , Citometria de Fluxo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/genética , Cavidade Peritoneal , Reação em Cadeia da Polimerase , Receptor CB2 de Canabinoide/genética
19.
Clin Investig Arterioscler ; 29(2): 51-59, 2017.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-28188022

RESUMO

INTRODUCTION: Oxidative stress and inflammation are determinant processes in the development of diabetic vascular complications. Heat shock protein 90 (HSP90) overexpression in atherosclerotic plaques plays a role in sustaining inflammatory mechanisms, and its specific inhibition prevents atherosclerosis. The present work investigates, in a mouse model of diabetes-driven atherosclerosis, whether atheroprotection by pharmacological HSP90 inhibition is accomplished by bolstering antioxidant defense mechanisms headed by nuclear factor erythroid-derived 2-like 2 (Nrf2). METHODS: Streptozotocin-induced diabetic apolipoprotein E-deficient mice were randomized to receive vehicle or HSP90 inhibitor (17-dimethylaminoethylamino-17-demethoxygeldanamycin, 4mg/kg) for 10 weeks. Aortic root sections were analyzed for plaque size and composition, transcription factor activity, and expression of inflammatory and antioxidant markers. In vitro studies were performed in murine macrophages cultured under hyperglycemic conditions. RESULTS: Treatment with HSP90 inhibitor promoted the activation of Nrf2 in the aortic tissue of diabetic mice (predominantly localized in macrophages and smooth muscle cells) and also in cultured cells. Nrf2 induction was associated with a concomitant inhibition of nuclear factor-κB (NF-κB) in atherosclerotic plaques, thus resulting in a significant reduction in lesion size and inflammatory component (leukocytes and cytokines). Furthermore, atheroprotection by HSP90 inhibition was linked to the induction of cytoprotective HSP70, antioxidant enzymes (heme oxygenase-1, superoxide dismutase and catalase) and autophagy machinery (LC3 and p62/SQSTM1) in aortic tissue. CONCLUSION: HSP90 inhibition protects from atherosclerosis in experimental diabetes through the induction of Nrf2-dependent cytoprotective mechanisms, reinforcing its therapeutic potential.


Assuntos
Aterosclerose/etiologia , Diabetes Mellitus Experimental/complicações , Proteínas de Choque Térmico HSP90/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Antioxidantes/metabolismo , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/fisiopatologia , Benzoquinonas/farmacologia , Inflamação/patologia , Lactamas Macrocíclicas/farmacologia , Macrófagos/metabolismo , Masculino , Camundongos , Estresse Oxidativo/fisiologia , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/patologia , Estreptozocina
20.
J Am Soc Nephrol ; 28(2): 575-585, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27609616

RESUMO

Diabetes is the main cause of CKD and ESRD worldwide. Chronic activation of Janus kinase and signal transducer and activator of transcription (STAT) signaling contributes to diabetic nephropathy by inducing genes involved in leukocyte infiltration, cell proliferation, and extracellular matrix accumulation. This study examined whether a cell-permeable peptide mimicking the kinase-inhibitory region of suppressor of cytokine signaling-1 (SOCS1) regulatory protein protects against nephropathy by suppressing STAT-mediated cell responses to diabetic conditions. In a mouse model combining hyperglycemia and hypercholesterolemia (streptozotocin diabetic, apoE-deficient mice), renal STAT activation status correlated with the severity of nephropathy. Notably, compared with administration of vehicle or mutant inactive peptide, administration of the SOCS1 peptidomimetic at either early or advanced stages of diabetes ameliorated STAT activity and resulted in reduced serum creatinine level, albuminuria, and renal histologic changes (mesangial expansion, tubular injury, and fibrosis) over time. Mice treated with the SOCS1 peptidomimetic also exhibited reduced kidney leukocyte recruitment (T lymphocytes and classic M1 proinflammatory macrophages) and decreased expression levels of proinflammatory and profibrotic markers that were independent of glycemic and lipid changes. In vitro, internalized peptide suppressed STAT activation and target gene expression induced by inflammatory and hyperglycemic conditions, reduced migration and proliferation in mesangial and tubuloepithelial cells, and altered the expression of cytokine-induced macrophage polarization markers. In conclusion, our study identifies SOCS1 mimicking as a feasible therapeutic strategy to halt the onset and progression of renal inflammation and fibrosis in diabetic kidney disease.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Peptidomiméticos/uso terapêutico , Proteína 1 Supressora da Sinalização de Citocina/uso terapêutico , Animais , Progressão da Doença , Masculino , Camundongos , Proteína 1 Supressora da Sinalização de Citocina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA