Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Neurobiol Dis ; 161: 105561, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34780863

RESUMO

Coronavirus disease 19 (COVID-19) is a respiratory illness caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). COVID-19 pathogenesis causes vascular-mediated neurological disorders via elusive mechanisms. SARS-CoV-2 infects host cells via the binding of viral Spike (S) protein to transmembrane receptor, angiotensin-converting enzyme 2 (ACE2). Although brain pericytes were recently shown to abundantly express ACE2 at the neurovascular interface, their response to SARS-CoV-2 S protein is still to be elucidated. Using cell-based assays, we found that ACE2 expression in human brain vascular pericytes was increased upon S protein exposure. Pericytes exposed to S protein underwent profound phenotypic changes associated with an elongated and contracted morphology accompanied with an enhanced expression of contractile and myofibrogenic proteins, such as α-smooth muscle actin (α-SMA), fibronectin, collagen I, and neurogenic locus notch homolog protein-3 (NOTCH3). On the functional level, S protein exposure promoted the acquisition of calcium (Ca2+) signature of contractile ensheathing pericytes characterized by highly regular oscillatory Ca2+ fluctuations. Furthermore, S protein induced lipid peroxidation, oxidative and nitrosative stress in pericytes as well as triggered an immune reaction translated by activation of nuclear factor-kappa-B (NF-κB) signaling pathway, which was potentiated by hypoxia, a condition associated with vascular comorbidities that exacerbate COVID-19 pathogenesis. S protein exposure combined to hypoxia enhanced the production of pro-inflammatory cytokines involved in immune cell activation and trafficking, namely macrophage migration inhibitory factor (MIF). Using transgenic mice expressing the human ACE2 that recognizes S protein, we observed that the intranasal infection with SARS-CoV-2 rapidly induced hypoxic/ischemic-like pericyte reactivity in the brain of transgenic mice, accompanied with an increased vascular expression of ACE2. Moreover, we found that SARS-CoV-2 S protein accumulated in the intranasal cavity reached the brain of mice in which the nasal mucosa is deregulated. Collectively, these findings suggest that SARS-CoV-2 S protein impairs the vascular and immune regulatory functions of brain pericytes, which may account for vascular-mediated brain damage. Our study provides a better understanding for the mechanisms underlying cerebrovascular disorders in COVID-19, paving the way to develop new therapeutic interventions.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Encéfalo/metabolismo , COVID-19/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia/metabolismo , Inflamação/metabolismo , Pericitos/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Actinas/metabolismo , Enzima de Conversão de Angiotensina 2/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/genética , Animais , Encéfalo/irrigação sanguínea , COVID-19/fisiopatologia , Sinalização do Cálcio , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Humanos , Hipóxia-Isquemia Encefálica/fisiopatologia , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/genética , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Miofibroblastos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Mucosa Nasal , Estresse Nitrosativo , Estresse Oxidativo , Pericitos/citologia , Pericitos/efeitos dos fármacos , Fenótipo , Receptor Notch3/metabolismo , Receptores de Coronavírus/efeitos dos fármacos , Receptores de Coronavírus/genética , Receptores de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/farmacologia
2.
Phytother Res ; 34(6): 1320-1328, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31840901

RESUMO

Ultraviolet (UV) radiation elicits melanogenesis and pigmentation in the skin. Apigenin (4',5,7-trihydroxyflavone [AGN]) is a plant flavone contained in various herbs, fruits, and vegetables. We herein investigated antimelanogenic properties of AGN and the molecular mechanisms of the action of AGN. In UVB-treated mice, AGN inhibited cutaneous hyperpigmentation and macrophage migration inhibitory factor (MIF) expression as a melanogenesis-related key factor. In mouse keratinocytes, AGN inhibited the expression of MIF and also the related factors (e.g., stem cell factor and proteinase-activated receptor 2) induced by MIF. In addition to ellagic acid as a casein kinase II (CK2) inhibitor, AGN suppressed CK2 enzymatic activity and UVB-induced CK2 expression and subsequent phosphorylation of IκB and MIF expression. These results suggest that AGN inhibits UVB-induced hyperpigmentation through the regulation of CK2-mediated MIF expression in keratinocytes.


Assuntos
Apigenina/fisiologia , Apigenina/uso terapêutico , Caseína Quinase II/efeitos dos fármacos , Hiperpigmentação/tratamento farmacológico , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Raios Ultravioleta/efeitos adversos , Animais , Apigenina/farmacologia , Humanos , Hiperpigmentação/patologia , Camundongos
3.
Alcohol Clin Exp Res ; 43(7): 1439-1451, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31009094

RESUMO

BACKGROUND: Macrophage migration inhibitory factor (MIF), a pluripotent immune regulator, is an emerging mediator in alcohol-related liver disease (ALD). MIF is associated with ALD progression through its chemokine- and cytokine-like activities. METHODS: Mechanistic studies into the role of MIF in ethanol (EtOH)-induced liver injury were performed in Mif-/- mice and in C57BL/6J mice treated with a small-molecule MIF antagonist, MIF098, after Gao-Binge (acute-on-chronic) EtOH feeding, an EtOH feeding protocol associated with hepatic neutrophilia and induction of the unfolded protein response (UPR). RESULTS: The MIF axis, for example, MIF and MIF receptors invariant polypeptide of major histocompatibility complex, class II antigen-associated (CD74), CXCR2, CXCR4, and CXCR7, was enhanced in the livers of alcoholic hepatitis (AH) patients as compared to healthy controls. Mif-/- mice were protected from hepatocellular injury after Gao-Binge feeding, independent of neutrophilia and inflammation, but were associated with the UPR. Interestingly, the UPR signature in AH patients and in mice following Gao-Binge feeding was biased toward cell death with increased expression of pro-cell death CCAAT-enhancer-binding protein homologous protein (CHOP) and decreased prosurvival GRP78. The UPR and liver injury 6 hours after binge were prevented both in Mif-/- mice and in MIF098-treated mice. However, both MIF interventions led to increased liver injury and exacerbated the hepatic UPR 9 hours after binge. Induction of upstream UPR signaling and expression of CHOP protein by thapsigargin in alpha mouse liver 12 hepatocytes were blunted by coexposure to MIF098, directly connecting MIF to UPR in hepatocytes. CONCLUSIONS: The current study revealed that, in addition to its cytokine/chemokine functions, MIF is an upstream regulator of UPR in response to EtOH feeding in mice. Importantly, both MIF and UPR can either protect or contribute to liver injury, dependent upon the stage or severity of EtOH-induced liver injury.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Oxirredutases Intramoleculares/efeitos dos fármacos , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/genética , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Benzoxazóis/farmacologia , Chaperona BiP do Retículo Endoplasmático , Feminino , Fator Estimulador de Colônias de Granulócitos/biossíntese , Interleucina-3/biossíntese , Oxirredutases Intramoleculares/antagonistas & inibidores , Fígado/efeitos dos fármacos , Fígado/patologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Proteínas Recombinantes de Fusão/biossíntese
4.
J Appl Oral Sci ; 27: e20180211, 2019 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-30810635

RESUMO

OBJECTIVE: The aim of this study was to evaluate the effects of gliclazide on oxidative stress, inflammation, and bone loss in an experimental periodontal disease model. MATERIAL AND METHODS: Male albino Wistar rats were divided into no ligature, ligature, and ligature with 1, 5, and 10 mg/kg gliclazide groups. Maxillae were fixed and scanned using micro-computed tomography to quantify linear and bone volume/tissue volume (BV/TV) and volumetric bone loss. Histopathological, immunohistochemical and immunofluorescence analyses were conducted to examine matrix metalloproteinase-2 (MMP-2), cyclooxygenase 2 (COX-2), cathepsin K, members of the receptor activator of the nuclear factor kappa-Β ligand (RANKL), receptor activator of nuclear factor kappa-Β (RANK), osteoprotegerin (OPG) pathway, macrophage migration inhibitory factor (MIF), superoxide dismutase-1 (SOD-1), glutathione peroxidase-1 (GPx-1), NFKB p 50 (Cytoplasm), NFKB p50 NLS (nuclear localization signal), PI3 kinase and AKT staining. Myeloperoxidase activity, malondialdehyde and glutathione levels, while interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) levels were evaluated by spectroscopic ultraviolet-visible analysis. A quantitative reverse transcription polymerase chain reaction was used to quantify the gene expression of the nuclear factor kappa B p50 subunit (NF-κB p50), phosphoinositide 3-kinase (PI3k), protein kinase B (AKT), and F4/80. RESULTS: Micro-computed tomography showed that the 1 mg/kg gliclazide treatment reduced linear bone loss compared to the ligature, 5 mg/kg gliclazide, and 10 mg/kg gliclazide treatments. All concentrations of gliclazide increased bone volume/tissue volume (BV/TV) compared to the ligature group. Treatment with 1 mg/kg gliclazide reduced myeloperoxidase activity, malondialdehyde, IL-1ß, and TNF-α levels (p≤0.05), and resulted in weak staining for COX-2, cathepsin k, MMP-2, RANK, RANKL, SOD-1, GPx-1,MIF and PI3k. In addition, down-regulation of NF-κB p50, PI3k, AKT, and F4/80 were observed, and OPG staining was strong after the 1 mg/kg gliclazide treatment. CONCLUSIONS: This treatment decreased neutrophil and macrophage migration, decreased the inflammatory response, and decreased bone loss in rats with ligature-induced periodontitis.


Assuntos
Perda do Osso Alveolar/tratamento farmacológico , Antioxidantes/farmacologia , Gliclazida/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Periodontite/tratamento farmacológico , Perda do Osso Alveolar/patologia , Animais , Antioxidantes/uso terapêutico , Catepsina K/análise , Imunofluorescência , Gengiva/química , Gengiva/patologia , Gliclazida/uso terapêutico , Glutationa/análise , Imuno-Histoquímica , Interleucina-1beta/análise , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Masculino , Malondialdeído/análise , Metaloproteinase 2 da Matriz/análise , Neutrófilos/efeitos dos fármacos , Periodontite/patologia , Peroxidase/análise , Ligante RANK/análise , Distribuição Aleatória , Ratos Wistar , Receptor Ativador de Fator Nuclear kappa-B/análise , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/análise , Microtomografia por Raio-X
5.
J Laryngol Otol ; 132(10): 891-895, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30270817

RESUMO

OBJECTIVES: This study aimed to compare serum macrophage migration inhibitory factor concentrations before and after oral steroid therapy in nasal polyps patients, and determine whether there is a difference between pre-treatment macrophage migration inhibitory factor concentrations and healthy individuals. METHODS: The study included 24 patients with nasal polyps and 25 healthy individuals. The patient group received 1 mg/kg oral steroid. RESULTS: The mean macrophage migration inhibitory factor concentration before oral steroid therapy was 3889.79 pg/ml in the patient group and 2334.52 pg/ml in the control group. Macrophage migration inhibitory factor concentrations were statistically significantly higher in the pre-oral steroid therapy patient group than in the control group (p = 0.017). The mean pre- and post-oral steroid therapy serum macrophage migration inhibitory factor concentrations were 3889.79 pg/ml and 2451.25 pg/ml, respectively. The reduction in macrophage migration inhibitory factor concentrations was statistically significant (p = 0.010). CONCLUSION: These findings suggest that concentrations of macrophage migration inhibitory factor may play a role in the pathogenesis of nasal polyps.


Assuntos
Glucocorticoides/administração & dosagem , Fatores Inibidores da Migração de Macrófagos/sangue , Metilprednisolona/administração & dosagem , Pólipos Nasais/diagnóstico , Pólipos Nasais/tratamento farmacológico , Administração Oral , Adulto , Estudos de Casos e Controles , Feminino , Hospitais Universitários , Humanos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Pólipos Nasais/sangue , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Método Simples-Cego , Tomografia Computadorizada por Raios X
6.
Head Neck ; 39(12): 2573-2584, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28963807

RESUMO

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in systemic, autoimmune, and inflammatory diseases, such as obesity, rheumatoid arthritis, and systemic lupus erythematosus. For the 2 past decades, MIF has been reported to participate in carcinogenesis, disease prognosis, tumor cell proliferation, invasion, and tumor-induced angiogenesis in many cancers. The purpose of this article is to review published experimental and clinical data for MIF and its involvement in upper aerodigestive tract cancers. Based on the current literature, we propose a biomolecular model describing the mechanisms underlying the involvement of MIF in the initiation, progression, apoptosis, and proliferation of head and neck tumor cells. In reference to this model, potential therapeutic approaches based on the use of MIF antagonists and neutralizing antibodies are described. It is concluded that MIF is a promising target for future therapeutic strategies, both with and without chemoradiation strategies.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias de Cabeça e Pescoço/patologia , Fatores Inibidores da Migração de Macrófagos/genética , Terapia de Alvo Molecular , Neovascularização Patológica/prevenção & controle , Apoptose/efeitos dos fármacos , Apoptose/genética , Biópsia por Agulha , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Progressão da Doença , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Humanos , Imuno-Histoquímica , Oxirredutases Intramoleculares/efeitos dos fármacos , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Sensibilidade e Especificidade , Células Tumorais Cultivadas
7.
Lupus ; 26(1): 62-72, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27230555

RESUMO

OBJECTIVE: Macrophage migration inhibitory factor (MIF) is a key regulator of both atherosclerosis and systemic lupus erythematosus (SLE), yet factors leading to its overproduction remain unclear. To explore regulation of MIF in SLE, we studied effects and potential mechanisms of type I interferon (IFN) and artesunate (ART), an antimalarial agent extracted from Chinese herbs, on levels of MIF. METHODS: Serum and peripheral blood cells from SLE patients and healthy controls were measured for MIF levels by ELISA and type I IFN-inducible gene expressions by real-time PCR, respectively, and assessed for associations by Spearman correlation. ART was added to human umbilical vein endothelial cell (HUVEC) cultures with or without prior IFNα-1b stimulation and to SLE peripheral blood mononuclear cell (PBMC) cultures. Protein levels of STATs and phosphorylated (p-) STATs in HUVECs were determined by Western blotting. RESULTS: Serum MIF levels were elevated in SLE patients and positively associated with disease activity (r = 0.86, p < 0.0001), accumulated damage (r = 0.34, p < 0.05), and IFN scores in SLE PBMCs (r = 0.74, p = 0.0002). The addition of IFNα-1b promoted MIF production in a time- and dose-dependent manner in HUVEC cultures. ART could inhibit expressions of IFN-inducible genes (LY6E and ISG15) in both HUVEC and SLE PBMC cultures, and suppress MIF production and over-expression of p-STAT1, but not p-STAT3 or STAT5, induced by IFNα-1b stimulation. IFNγ-induced expression of p-STAT1 in HUVECs was not inhibited by ART. CONCLUSION: MIF could be regulated by type I IFN in SLE patients. ART counteracts the effect of IFNα to inhibit MIF production by blocking STAT1 phosphorylation and thus may have therapeutic potential for SLE-associated atherosclerosis.


Assuntos
Artemisininas/farmacologia , Oxirredutases Intramoleculares/metabolismo , Lúpus Eritematoso Sistêmico/fisiopatologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fator de Transcrição STAT1/metabolismo , Adulto , Antimaláricos/administração & dosagem , Antimaláricos/farmacologia , Artemisininas/administração & dosagem , Artesunato , Western Blotting , Estudos de Casos e Controles , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Interferon Tipo I/metabolismo , Interferon-alfa/administração & dosagem , Interferon-alfa/metabolismo , Oxirredutases Intramoleculares/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT1/genética , Fatores de Tempo , Adulto Jovem
8.
Med Res Rev ; 36(3): 440-60, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26777977

RESUMO

Strong evidence has been presented linking chronic inflammation to the onset and pathogenesis of cancer. The multifunctional pro-inflammatory protein macrophage migration inhibitory factor (MIF) occupies a central role in the inflammatory pathway and has been implicated in the tumorigenesis, angiogenesis, and metastasis of many cancer phenotypes. This review highlights the current state of the art, which presents MIF, and the second member of the MIF structural superfamily, D-DT (MIF2), as significant mediators in the inflammatory-cancer axis. Although the mechanism by which MIF asserts its biological activity has yet to be fully understood, it has become clear in recent years that for certain phenotypes of cancer, MIF represents a valid therapeutic target. Current research efforts have focused on small molecule approaches that target MIF's unique tautomerase active site and neutralization of MIF with anti-MIF antibodies. These approaches have yielded promising results in a number of preclinical murine cancer models and have helped to increase our understanding of MIF biological activity. More recently, MIF's involvement in a number of key protein-protein interactions, such as with CD74 and HSP90, has been highlighted and provides a novel platform for the development of anti-MIF chemotherapeutic strategies in the future.


Assuntos
Oxirredutases Intramoleculares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Neoplasias/terapia , Humanos , Oxirredutases Intramoleculares/química , Fatores Inibidores da Migração de Macrófagos/química , Neoplasias/metabolismo
9.
Braz J Med Biol Res ; 46(9): 746-51, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24036910

RESUMO

Macrophage migration inhibitory factor (MIF), a pleiotropic cytokine, plays an important role in the pathogenesis of atrial fibrillation; however, the upstream regulation of MIF in atrial myocytes remains unclear. In the present study, we investigated whether and how MIF is regulated in response to the renin-angiotensin system and oxidative stress in atrium myocytes (HL-1 cells). MIF protein and mRNA levels in HL-1 cells were assayed using immunofluorescence, real-time PCR, and Western blot. The result indicated that MIF was expressed in the cytoplasm of HL-1 cells. Hydrogen peroxide (H2O2), but not angiotensin II, stimulated MIF expression in HL-1 cells. H2O2-induced MIF protein and gene levels increased in a dose-dependent manner and were completely abolished in the presence of catalase. H2O2-induced MIF production was completely inhibited by tyrosine kinase inhibitors genistein and PP1, as well as by protein kinase C (PKC) inhibitor GF109203X, suggesting that redox-sensitive MIF production is mediated through tyrosine kinase and PKC-dependent mechanisms in HL-1 cells. These results suggest that MIF is upregulated by HL-1 cells in response to redox stress, probably by the activation of Src and PKC.


Assuntos
Peróxido de Hidrogênio/farmacologia , Oxirredutases Intramoleculares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxidantes/farmacologia , Proteína Quinase C/metabolismo , Quinases da Família src/metabolismo , Angiotensina II/metabolismo , Animais , Western Blotting , Linhagem Celular , Imuno-Histoquímica , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos , Microscopia Confocal , Estresse Oxidativo/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Sistema Renina-Angiotensina/fisiologia
10.
Braz. j. med. biol. res ; 46(9): 746-751, 19/set. 2013. graf
Artigo em Inglês | LILACS | ID: lil-686569

RESUMO

Macrophage migration inhibitory factor (MIF), a pleiotropic cytokine, plays an important role in the pathogenesis of atrial fibrillation; however, the upstream regulation of MIF in atrial myocytes remains unclear. In the present study, we investigated whether and how MIF is regulated in response to the renin-angiotensin system and oxidative stress in atrium myocytes (HL-1 cells). MIF protein and mRNA levels in HL-1 cells were assayed using immunofluorescence, real-time PCR, and Western blot. The result indicated that MIF was expressed in the cytoplasm of HL-1 cells. Hydrogen peroxide (H2O2), but not angiotensin II, stimulated MIF expression in HL-1 cells. H2O2-induced MIF protein and gene levels increased in a dose-dependent manner and were completely abolished in the presence of catalase. H2O2-induced MIF production was completely inhibited by tyrosine kinase inhibitors genistein and PP1, as well as by protein kinase C (PKC) inhibitor GF109203X, suggesting that redox-sensitive MIF production is mediated through tyrosine kinase and PKC-dependent mechanisms in HL-1 cells. These results suggest that MIF is upregulated by HL-1 cells in response to redox stress, probably by the activation of Src and PKC.


Assuntos
Animais , Camundongos , Peróxido de Hidrogênio/farmacologia , Oxirredutases Intramoleculares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxidantes/farmacologia , Proteína Quinase C/metabolismo , Quinases da Família src/metabolismo , Angiotensina II/metabolismo , Western Blotting , Linhagem Celular , Imuno-Histoquímica , Oxirredutases Intramoleculares/genética , Microscopia Confocal , Fatores Inibidores da Migração de Macrófagos/genética , Estresse Oxidativo/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Sistema Renina-Angiotensina/fisiologia
11.
J Periodontal Res ; 48(4): 527-32, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23298274

RESUMO

BACKGROUND AND OBJECTIVE: Macrophage migration-inhibitory factor (MIF) plays crucial roles in the recruitment and activation of macrophages as well as in helping to kill bacteria. This study investigated the expression profile of MIF in human gingiva under different periodontal conditions and its expression patterns induced by Porphyromonas gingivalis lipopolysaccharide (LPS) in gingival epithelia. MATERIAL AND METHODS: Gingival tissue samples were collected from deep pockets and clinically healthy sites of 22 nonsmoking subjects with chronic periodontitis. The expression of MIF mRNA and protein was evaluated using real-time PCR and immunohistochemistry, respectively. The in vitro study analyzed the effects of P. gingivalis LPS on the expression of MIF in a reconstituted human gingival epithelia (RHGE) model. RESULTS: In gingival epithelia, MIF protein was diffusely expressed from the basal layer to the granular and spinous layers; whereas, in the underlying connective tissues, MIF was observed around the dilated blood vessels in the deep-pocket tissues. A significantly lower level of expression of MIF mRNA and an increased level of expression of MIF protein were found in deep-pocket tissues compared with clinically healthy tissues. Expression of MIF mRNA in the RHGE model was significantly down-regulated by P. gingivalis LPS. CONCLUSION: The present study suggests that MIF expression may be related to periodontal conditions and that its expression profile could be modulated by P. gingivalis LPS. MIF may play a role in periodontal pathogenesis.


Assuntos
Gengiva/patologia , Oxirredutases Intramoleculares/análise , Lipopolissacarídeos/farmacologia , Fatores Inibidores da Migração de Macrófagos/análise , Porphyromonas gingivalis/metabolismo , Adulto , Capilares/patologia , Periodontite Crônica/patologia , Tecido Conjuntivo/irrigação sanguínea , Tecido Conjuntivo/patologia , Epitélio/efeitos dos fármacos , Epitélio/patologia , Escherichia coli/metabolismo , Gengiva/efeitos dos fármacos , Humanos , Oxirredutases Intramoleculares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Pessoa de Meia-Idade , Bolsa Periodontal/patologia , Técnicas de Cultura de Tecidos
13.
Br J Dermatol ; 166(3): 653-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22050122

RESUMO

BACKGROUND: Recently, evidence has been obtained to suggest that inflammation is provoked through upregulation of macrophage migration inhibitory factor (MIF) expression by steroids. However, little is known regarding the effect of steroids on MIF expression in human keratinocytes and the counter-effect of epigallocatechin-3-gallate (EGCG), a member of the class of green tea polyphenols. OBJECTIVES: We determined whether or not steroids cause the upregulation of MIF in human keratinocytes, and if so, whether or not EGCG suppresses MIF upregulation in keratinocytes by steroids. We then assessed the effects of EGCG on MIF-induced Th-related chemokine and cytokine expression in keratinocytes. METHODS: HaCaT keratinocytes were first treated with dexamethasone in the presence or absence of EGCG in the culture medium. The keratinocytes were then treated with recombinant human (rh)-MIF in the presence or absence of EGCG in the culture medium. The expression of mRNA and protein in Th-related cytokines and chemokines, including MIF in the keratinocytes, was measured by real-time reverse transcription-polymerase chain reaction, Western blotting and enzyme-linked immunosorbent assay. RESULTS: Dexamethasone significantly enhanced MIF expression in human keratinocytes, and EGCG significantly downregulated the expression of dexamethasone-induced MIF. EGCG also significantly downregulated rh-MIF-induced expression of Th-related cytokines and chemokines, such as interleukin (IL)-6, IL-18, transforming growth factor-ß, CCL17, CCL22 and CXCL10, in human keratinocytes. CONCLUSIONS: These results demonstrated that EGCG may have a novel pharmacological effect to prevent steroid-induced tachyphylaxis and inflammation by suppressing the expression of MIF in human keratinocytes.


Assuntos
Camellia sinensis , Catequina/análogos & derivados , Queratinócitos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Anti-Inflamatórios/farmacologia , Catequina/farmacologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Dermatite/tratamento farmacológico , Dexametasona/farmacologia , Resistência a Medicamentos , Glucocorticoides/farmacologia , Humanos , Queratinócitos/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Recombinantes , Regulação para Cima
14.
Phytother Res ; 25(5): 744-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21425378

RESUMO

The protective effects of plant polyphenol intake on cardiovascular morbidity and mortality are widely acknowledged. Caffeine-free chicory coffee is a rich source of plant phenolics, including caffeic acid, which inhibits in vitro platelet aggregation, and also phenylpyruvate tautomerase enzymatic activity of the proinflammatory cytokine, macrophage migration inhibitory factor (MIF). To assess whether chicory coffee consumption might confer cardiovascular benefits a clinical intervention study was performed with 27 healthy volunteers, who consumed 300 mL chicory coffee every day for 1 week. The dietary intervention produced variable effects on platelet aggregation, depending on the inducer used for the aggregation test. Whole blood and plasma viscosity were both significantly decreased, along with serum MIF levels, after 1 week of chicory coffee consumption. Moreover, significant improvements were seen in red blood cell deformability. No changes in hematocrit, fibrinogen level or red blood cell counts were detected. The full spectrum of these effects is unlikely to be attributable to a single compound present in chicory coffee, nevertheless, the phenolics, including caffeic acid, are expected to play a substantial role. In conclusion, our study offers an encouraging starting-point to delineate the antithrombotic and antiinflammatory effects of phenolic compounds found in chicory coffee.


Assuntos
Antioxidantes/farmacologia , Ácidos Cafeicos/farmacologia , Cichorium intybus/química , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Trombose/prevenção & controle , Plaquetas/efeitos dos fármacos , Viscosidade Sanguínea/efeitos dos fármacos , Deformação Eritrocítica/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Comportamento Alimentar , Feminino , Humanos , Oxirredutases Intramoleculares/sangue , Oxirredutases Intramoleculares/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/sangue , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Masculino , Raízes de Plantas/química , Agregação Plaquetária/efeitos dos fármacos , Adulto Jovem
15.
Inflamm Allergy Drug Targets ; 9(1): 10-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19906008

RESUMO

Primary Sjögren's syndrome (pSS) is a common autoimmune disease which can lead to considerable complications and diminished quality of life. Recent insights into disease mechanisms and the advent of biological agents have provided new options for the treatment of pSS. In particular, B cell targeted intervention has shown promising results. In this review, we focus on emerging treatment strategies and therapeutic targets beyond B cells. Interference with proinflammatory cytokines and mechanisms that link innate and adaptive immunity offers new options in the treatment of pSS. Approaches directed against interleukin (IL)-1beta, Toll-like receptors and the inflammasome are emerging. Targeting IL-12, IL-18, the IL-23/IL-17 system, macrophage migration inhibitory factor and chemokines might be considered. The inhibition of apoptosis of glandular cells, the promotion of cell regeneration and organ-specific stem cell transplantation are potential strategies directed at preserving and restoring functional exocrine tissue. The recognition of patients who benefit most from a particular strategy might help to design more efficient therapeutic approaches. Since efficacy of many agents depends on the presence of residual functional glandular tissue, future studies should focus on patients with recent onset of pSS.


Assuntos
Síndrome de Sjogren/tratamento farmacológico , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Quimiocinas/efeitos dos fármacos , Quimiocinas/imunologia , Glucocorticoides/uso terapêutico , Humanos , Hidroxicloroquina/uso terapêutico , Imunossupressores/uso terapêutico , Interleucina-12/imunologia , Interleucina-17/imunologia , Interleucina-18/imunologia , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/imunologia , Interleucina-23/imunologia , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/imunologia , Síndrome de Sjogren/imunologia , Transplante de Células-Tronco , Receptores Toll-Like/efeitos dos fármacos , Receptores Toll-Like/imunologia
16.
Crit Care ; 13(4): 170, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19678905

RESUMO

Bacterial meningitis is a complex disorder in which injury is caused, in part, by the causative organism and, in part, by the host's own inflammatory response. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine and a neuro-endocrine mediator that might play a role in pneumococcal meningitis. Here, we discuss the role of MIF in infection, the brain, and corticosteroids and conclude that experimental meningitis studies have to determine whether MIF is a potential target for adjunctive therapy in pneumococcal meningitis.


Assuntos
Corticosteroides/uso terapêutico , Encefalite/imunologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Corticosteroides/administração & dosagem , Encefalite/tratamento farmacológico , Encefalite/fisiopatologia , Humanos , Fatores Inibidores da Migração de Macrófagos/líquido cefalorraquidiano , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos
17.
Lancet ; 373(9678): 1905-17, 2009 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-19482216

RESUMO

Glucocorticoid resistance or insensitivity is a major barrier to the treatment of several common inflammatory diseases-including chronic obstructive pulmonary disease and acute respiratory distress syndrome; it is also an issue for some patients with asthma, rheumatoid arthritis, and inflammatory bowel disease. Several molecular mechanisms of glucocorticoid resistance have now been identified, including activation of mitogen-activated protein (MAP) kinase pathways by certain cytokines, excessive activation of the transcription factor activator protein 1, reduced histone deacetylase-2 (HDAC2) expression, raised macrophage migration inhibitory factor, and increased P-glycoprotein-mediated drug efflux. Patients with glucocorticoid resistance can be treated with alternative broad-spectrum anti-inflammatory treatments, such as calcineurin inhibitors and other immunomodulators, or novel anti-inflammatory treatments, such as inhibitors of phosphodiesterase 4 or nuclear factor kappaB, although these drugs are all likely to have major side-effects. An alternative treatment strategy is to reverse glucocorticoid resistance by blocking its underlying mechanisms. Some examples of this approach are inhibition of p38 MAP kinase, use of vitamin D to restore interleukin-10 response, activation of HDAC2 expression by use of theophylline, antioxidants, or phosphoinositide-3-kinase-delta inhibitors, and inhibition of macrophage migration inhibitory factor and P-glycoprotein.


Assuntos
Anti-Inflamatórios/uso terapêutico , Resistência a Medicamentos , Glucocorticoides/uso terapêutico , Inflamação/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/efeitos dos fármacos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/imunologia , Asma/tratamento farmacológico , Inibidores de Calcineurina , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Resistência a Medicamentos/imunologia , Predisposição Genética para Doença/genética , Histona Desacetilase 2 , Histona Desacetilases/efeitos dos fármacos , Histona Desacetilases/genética , Histona Desacetilases/imunologia , Humanos , Inflamação/imunologia , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/imunologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/imunologia , NF-kappa B/antagonistas & inibidores , Inibidores da Fosfodiesterase 4 , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Proteínas Repressoras/efeitos dos fármacos , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Fator de Transcrição AP-1/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/imunologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Ativação Transcricional/imunologia
18.
J Ethnopharmacol ; 120(2): 259-63, 2008 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18835428

RESUMO

AIM OF THIS STUDY: Daphnetin was regarded as the mark compound for quality control of Zushima-Pian, a traditional Chinese medicine tablet for treating rheumatoid arthritis (RA). However, no in vivo study on the therapeutic effects of daphnetin for RA has been reported. MATERIALS AND METHODS: The adjuvant arthritic (AA) rat model was developed to evaluate the anti-arthritic effects of daphnetin. After immunized with Freund's complete adjuvant (FCA), the rats were treated with daphnetin (2.25 and 4.5mg/kg) for three weeks. We determined the change of secondary paw swelling and the arthritis scores of these tested rats. The severity of arthritis in the knee joints was evaluated by histological assessment of cartilage destruction. The levels of IL-1, TNF-alpha and MIF in the serum were measured by ELISA. RESULTS AND CONCLUSIONS: Our results showed that daphnetin significantly reduced paw swelling and decreased the arthritis scores. The pathological examination demonstrated that articular cartilage degeneration with synovial hyperplasia and inflammatory cells infiltration in AA rats were suppressed by daphnetin. There was significant reduction in production of interleukin-1 (IL-1), tumor necrosis factor (TNF-alpha) and macrophage migration inhibitory factor (MIF) in serum of AA rats treated with daphnetin except the low dose group (2.25mg/kg) on TNF-alpha. In conclusion, we demonstrates that daphnetin is highly effective on preventing and suppressing the development and progression of adjuvant-induced arthritis and provides direct evidences that daphnetin is one of the active principle of Zushima-Pian for treating rheumatoid arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Umbeliferonas/farmacologia , Animais , Artrite Experimental/fisiopatologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/fisiopatologia , Progressão da Doença , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/farmacologia , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund , Interleucina-1/metabolismo , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Umbeliferonas/administração & dosagem
19.
Immunol Cell Biol ; 86(3): 232-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18040285

RESUMO

Macrophage migration inhibitory factor (MIF) has an amazing history of rediscoveries and controversies surroundings its true biological function. It has been classified as a powerful cytokine capable of inducing tumour necrosis factor (TNF)-alpha, IL-1beta, IL-6, IL-8, PGE2 along with its ability to override glucocorticoid activity in relation to TNF-alpha release from monocytes. However, our recent study has failed to reproduce findings on MIF as a factor with cytokine-inducing properties but it has confirmed that MIF is capable of inducing glucocorticoid-counter regulating activity and amplifying LPS-driven cytokine responses. The aim of this review is to analyse the plethora of data surrounding MIF not just as a cytokine, but also as a hormone-like molecule, enzyme with atypical properties and as a thioredoxin-like protein to address fundamental questions about MIF functionality.


Assuntos
Oxirredutases Intramoleculares/imunologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Oxirredução , Linfócitos T/enzimologia , Acetaminofen/administração & dosagem , Acetaminofen/análogos & derivados , Animais , Movimento Celular/imunologia , Suscetibilidade a Doenças/enzimologia , Retroalimentação Fisiológica , Glucocorticoides/imunologia , Glucocorticoides/metabolismo , Humanos , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/genética , Polimorfismo Genético , Substâncias Redutoras/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...