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

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Encéfalo/metabolismo , COVID-19/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia/metabolismo , Inflamación/metabolismo , Pericitos/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Actinas/metabolismo , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/genética , Animales , Encéfalo/irrigación sanguínea , COVID-19/fisiopatología , Señalización del Calcio , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Humanos , Hipoxia-Isquemia Encefálica/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Miofibroblastos , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Mucosa Nasal , Estrés Nitrosativo , Estrés Oxidativo , Pericitos/citología , Pericitos/efectos de los fármacos , Fenotipo , Receptor Notch3/metabolismo , Receptores de Coronavirus/efectos de los fármacos , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/farmacología
2.
Phytother Res ; 34(6): 1320-1328, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31840901

RESUMEN

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.


Asunto(s)
Apigenina/fisiología , Apigenina/uso terapéutico , Quinasa de la Caseína II/efectos de los fármacos , Hiperpigmentación/tratamiento farmacológico , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Animales , Apigenina/farmacología , Humanos , Hiperpigmentación/patología , Ratones
3.
Alcohol Clin Exp Res ; 43(7): 1439-1451, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31009094

RESUMEN

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.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Oxidorreductasas Intramoleculares/efectos de los fármacos , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/genética , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Benzoxazoles/farmacología , Chaperón BiP del Retículo Endoplásmico , Femenino , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Interleucina-3/biosíntesis , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Hígado/efectos de los fármacos , Hígado/patología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Proteínas Recombinantes de Fusión/biosíntesis
4.
J Appl Oral Sci ; 27: e20180211, 2019 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-30810635

RESUMEN

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.


Asunto(s)
Pérdida de Hueso Alveolar/tratamiento farmacológico , Antioxidantes/farmacología , Gliclazida/farmacología , Estrés Oxidativo/efectos de los fármacos , Periodontitis/tratamiento farmacológico , Pérdida de Hueso Alveolar/patología , Animales , Antioxidantes/uso terapéutico , Catepsina K/análisis , Técnica del Anticuerpo Fluorescente , Encía/química , Encía/patología , Gliclazida/uso terapéutico , Glutatión/análisis , Inmunohistoquímica , Interleucina-1beta/análisis , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Masculino , Malondialdehído/análisis , Metaloproteinasa 2 de la Matriz/análisis , Neutrófilos/efectos de los fármacos , Periodontitis/patología , Peroxidasa/análisis , Ligando RANK/análisis , Distribución Aleatoria , Ratas Wistar , Receptor Activador del Factor Nuclear kappa-B/análisis , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/análisis , Microtomografía por Rayos X
5.
J Laryngol Otol ; 132(10): 891-895, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30270817

RESUMEN

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.


Asunto(s)
Glucocorticoides/administración & dosificación , Factores Inhibidores de la Migración de Macrófagos/sangre , Metilprednisolona/administración & dosificación , Pólipos Nasales/diagnóstico , Pólipos Nasales/tratamiento farmacológico , Administración Oral , Adulto , Estudios de Casos y Controles , Femenino , Hospitales Universitarios , Humanos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Pólipos Nasales/sangre , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad , Método Simple Ciego , Tomografía Computarizada por Rayos X
6.
Head Neck ; 39(12): 2573-2584, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28963807

RESUMEN

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.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de Cabeza y Cuello/patología , Factores Inhibidores de la Migración de Macrófagos/genética , Terapia Molecular Dirigida , Neovascularización Patológica/prevención & control , Apoptosis/efectos de los fármacos , Apoptosis/genética , Biopsia con Aguja , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Humanos , Inmunohistoquímica , Oxidorreductasas Intramoleculares/efectos de los fármacos , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Sensibilidad y Especificidad , Células Tumorales Cultivadas
7.
Lupus ; 26(1): 62-72, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27230555

RESUMEN

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.


Asunto(s)
Artemisininas/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Lupus Eritematoso Sistémico/fisiopatología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factor de Transcripción STAT1/metabolismo , Adulto , Antimaláricos/administración & dosificación , Antimaláricos/farmacología , Artemisininas/administración & dosificación , Artesunato , Western Blotting , Estudios de Casos y Controles , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interferón Tipo I/metabolismo , Interferón-alfa/administración & dosificación , Interferón-alfa/metabolismo , Oxidorreductasas Intramoleculares/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico/tratamiento farmacológico , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/genética , Factores de Tiempo , Adulto Joven
8.
Med Res Rev ; 36(3): 440-60, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26777977

RESUMEN

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.


Asunto(s)
Oxidorreductasas Intramoleculares/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Neoplasias/terapia , Humanos , Oxidorreductasas Intramoleculares/química , Factores Inhibidores de la Migración de Macrófagos/química , Neoplasias/metabolismo
9.
Braz J Med Biol Res ; 46(9): 746-51, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24036910

RESUMEN

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.


Asunto(s)
Peróxido de Hidrógeno/farmacología , Oxidorreductasas Intramoleculares/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidantes/farmacología , Proteína Quinasa C/metabolismo , Familia-src Quinasas/metabolismo , Angiotensina II/metabolismo , Animales , Western Blotting , Línea Celular , Inmunohistoquímica , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Microscopía Confocal , Estrés Oxidativo/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Sistema Renina-Angiotensina/fisiología
10.
Braz. j. med. biol. res ; 46(9): 746-751, 19/set. 2013. graf
Artículo en Inglés | LILACS | ID: lil-686569

RESUMEN

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.


Asunto(s)
Animales , Ratones , Peróxido de Hidrógeno/farmacología , Oxidorreductasas Intramoleculares/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxidantes/farmacología , Proteína Quinasa C/metabolismo , Familia-src Quinasas/metabolismo , Angiotensina II/metabolismo , Western Blotting , Línea Celular , Inmunohistoquímica , Oxidorreductasas Intramoleculares/genética , Microscopía Confocal , Factores Inhibidores de la Migración de Macrófagos/genética , Estrés Oxidativo/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Sistema Renina-Angiotensina/fisiología
11.
J Periodontal Res ; 48(4): 527-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23298274

RESUMEN

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.


Asunto(s)
Encía/patología , Oxidorreductasas Intramoleculares/análisis , Lipopolisacáridos/farmacología , Factores Inhibidores de la Migración de Macrófagos/análisis , Porphyromonas gingivalis/metabolismo , Adulto , Capilares/patología , Periodontitis Crónica/patología , Tejido Conectivo/irrigación sanguínea , Tejido Conectivo/patología , Epitelio/efectos de los fármacos , Epitelio/patología , Escherichia coli/metabolismo , Encía/efectos de los fármacos , Humanos , Oxidorreductasas Intramoleculares/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Persona de Mediana Edad , Bolsa Periodontal/patología , Técnicas de Cultivo de Tejidos
13.
Br J Dermatol ; 166(3): 653-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22050122

RESUMEN

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.


Asunto(s)
Camellia sinensis , Catequina/análogos & derivados , Queratinocitos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Polifenoles/farmacología , Antiinflamatorios/farmacología , Catequina/farmacología , Quimiocinas/metabolismo , Citocinas/metabolismo , Dermatitis/tratamiento farmacológico , Dexametasona/farmacología , Resistencia a Medicamentos , Glucocorticoides/farmacología , Humanos , Queratinocitos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas Recombinantes , Regulación hacia Arriba
14.
Phytother Res ; 25(5): 744-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21425378

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Ácidos Cafeicos/farmacología , Cichorium intybus/química , Extractos Vegetales/farmacología , Polifenoles/farmacología , Trombosis/prevención & control , Plaquetas/efectos de los fármacos , Viscosidad Sanguínea/efectos de los fármacos , Deformación Eritrocítica/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Conducta Alimentaria , Femenino , Humanos , Oxidorreductasas Intramoleculares/sangre , Oxidorreductasas Intramoleculares/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/sangre , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Masculino , Raíces de Plantas/química , Agregación Plaquetaria/efectos de los fármacos , Adulto Joven
15.
Inflamm Allergy Drug Targets ; 9(1): 10-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19906008

RESUMEN

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.


Asunto(s)
Síndrome de Sjögren/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Quimiocinas/efectos de los fármacos , Quimiocinas/inmunología , Glucocorticoides/uso terapéutico , Humanos , Hidroxicloroquina/uso terapéutico , Inmunosupresores/uso terapéutico , Interleucina-12/inmunología , Interleucina-17/inmunología , Interleucina-18/inmunología , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Interleucina-23/inmunología , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/inmunología , Síndrome de Sjögren/inmunología , Trasplante de Células Madre , Receptores Toll-Like/efectos de los fármacos , Receptores Toll-Like/inmunología
16.
Crit Care ; 13(4): 170, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19678905

RESUMEN

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.


Asunto(s)
Corticoesteroides/uso terapéutico , Encefalitis/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Corticoesteroides/administración & dosificación , Encefalitis/tratamiento farmacológico , Encefalitis/fisiopatología , Humanos , Factores Inhibidores de la Migración de Macrófagos/líquido cefalorraquídeo , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos
17.
Lancet ; 373(9678): 1905-17, 2009 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-19482216

RESUMEN

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.


Asunto(s)
Antiinflamatorios/uso terapéutico , Resistencia a Medicamentos , Glucocorticoides/uso terapéutico , Inflamación/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/inmunología , Asma/tratamiento farmacológico , Inhibidores de la Calcineurina , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Resistencia a Medicamentos/inmunología , Predisposición Genética a la Enfermedad/genética , Histona Desacetilasa 2 , Histona Desacetilasas/efectos de los fármacos , Histona Desacetilasas/genética , Histona Desacetilasas/inmunología , Humanos , Inflamación/inmunología , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/inmunología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , FN-kappa B/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa 4 , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Proteínas Represoras/efectos de los fármacos , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Factor de Transcripción AP-1/efectos de los fármacos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Activación Transcripcional/inmunología
18.
J Ethnopharmacol ; 120(2): 259-63, 2008 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-18835428

RESUMEN

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.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Umbeliferonas/farmacología , Animales , Artritis Experimental/fisiopatología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Ensayo de Inmunoadsorción Enzimática , Adyuvante de Freund , Interleucina-1/metabolismo , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Umbeliferonas/administración & dosificación
19.
Immunol Cell Biol ; 86(3): 232-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18040285

RESUMEN

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.


Asunto(s)
Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Oxidación-Reducción , Linfocitos T/enzimología , Acetaminofén/administración & dosificación , Acetaminofén/análogos & derivados , Animales , Movimiento Celular/inmunología , Susceptibilidad a Enfermedades/enzimología , Retroalimentación Fisiológica , Glucocorticoides/inmunología , Glucocorticoides/metabolismo , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/genética , Polimorfismo Genético , Sustancias Reductoras/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...