Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Mult Scler ; 30(1): 35-43, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37982154

RESUMEN

BACKGROUND: Macrophage migration inhibitory factor (MIF) is a cytokine linked to multiple sclerosis (MS) progression that is thought to be inhibited by ibudilast. SPRINT-MS was a phase 2 placebo-controlled trial of ibudilast in progressive multiple sclerosis (PMS). OBJECTIVE: To determine whether baseline MIF levels predict imaging outcomes and assess the effects of ibudilast on serum and cerebrospinal fluid (CSF) MIF levels in people with PMS treated with ibudilast. METHODS: Participants in the SPRINT-MS trial were treated with either ibudilast or placebo and underwent brain magnetic resonance imaging (MRI) every 24 weeks over a duration of 96 weeks. MIF was measured in serum and CSF. RESULTS: MIF levels were compared with imaging outcomes in 223 participants from the SPRINT-MS study. In the primary progressive multiple sclerosis (PPMS) cohort, males had higher serum (p < 0.001) and CSF (p = 0.01) MIF levels, as compared with females. Higher baseline serum MIF levels in PPMS were associated with faster brain atrophy (beta = -0.113%, 95% confidence interval (CI): -0.204% to -0.021%; p = 0.016). These findings were not observed in secondary progressive multiple sclerosis (SPMS). Ibudilast did not affect either serum or CSF MIF levels. CONCLUSIONS: Serum MIF levels were associated with male sex and predicted brain atrophy in PPMS, but not SPMS. Ibudilast did not demonstrate an effect on MIF levels, as compared with placebo, although we cannot exclude a functional effect.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Factores Inhibidores de la Migración de Macrófagos , Esclerosis Múltiple Crónica Progresiva , Esclerosis Múltiple , Femenino , Humanos , Masculino , Atrofia/patología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Factores Inhibidores de la Migración de Macrófagos/líquido cefalorraquídeo , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple Crónica Progresiva/diagnóstico por imagen , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Esclerosis Múltiple Crónica Progresiva/patología
2.
CNS Neurosci Ther ; 29(12): 3802-3814, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37334735

RESUMEN

BACKGROUND: Macrophage migration inhibitory factor (MIF) is an important mediator of neuropathology in various central nervous system (CNS) diseases. However, little is known about its inducers for production from the nerve cells, as well as the underlying regulatory mechanism. Injury-induced HIF-1α has been shown to exacerbate neuroinflammation by activating multiple downstream target molecules. It is postulated that HIF-1α is involved in the regulation of MIF following spinal cord injury (SCI). METHODS: SCI model of Sprague-Dawley rats was established by cord contusion at T8-T10. The dynamic changes of HIF-1α and MIF protein levels at lesion site of rat spinal cord were determined by Western blot. The specific cell types of HIF-1α and MIF expression were examined by immunostaining. Primary astrocytes were isolated from the spinal cord, cultured and stimulated with various agonist or inhibitor of HIF-1α for analysis of HIF-1α-mediated expression of MIF. Luciferase report assay was used to determine the relationship between HIF-1α and MIF. The Basso, Beattie, and Bresnahan (BBB) locomotor scale was used to assess the locomotor function following SCI. RESULTS: The protein levels of HIF-1α and MIF at lesion site were significantly elevated by SCI. Immunofluorescence demonstrated that both HIF-1α and MIF were abundantly expressed in the astrocytes of the spinal cord. By using various agonists or inhibitors of HIF-1α, it was shown that HIF-1α sufficiently induced astrocytic production of MIF. Mechanistically, HIF-1α promoted MIF expression through interaction with MIF promoter. Inhibition of HIF-1α activity using specific inhibitor markedly reduced the protein levels of MIF at lesion site following SCI, which in turn favored for the functional recovery. CONCLUSION: SCI-induced activation of HIF-1α is able to promote MIF production from astrocytes. Our results have provided new clues for SCI-induced production of DAMPs, which may be helpful for clinical treatment of neuroinflammation.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos , Traumatismos de la Médula Espinal , Ratas , Animales , Ratas Sprague-Dawley , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/farmacología , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias , Traumatismos de la Médula Espinal/patología , Médula Espinal/metabolismo , Recuperación de la Función
3.
Thorax ; 78(7): 661-673, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36344253

RESUMEN

BACKGROUND: Severe neutrophilic asthma is resistant to treatment with glucocorticoids. The immunomodulatory protein macrophage migration inhibitory factor (MIF) promotes neutrophil recruitment to the lung and antagonises responses to glucocorticoids. We hypothesised that MIF promotes glucocorticoid resistance of neutrophilic inflammation in severe asthma. METHODS: We examined whether sputum MIF protein correlated with clinical and molecular characteristics of severe neutrophilic asthma in the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort. We also investigated whether MIF regulates neutrophilic inflammation and glucocorticoid responsiveness in a murine model of severe asthma in vivo. RESULTS: MIF protein levels positively correlated with the number of exacerbations in the previous year, sputum neutrophils and oral corticosteroid use across all U-BIOPRED subjects. Further analysis of MIF protein expression according to U-BIOPRED-defined transcriptomic-associated clusters (TACs) revealed increased MIF protein and a corresponding decrease in annexin-A1 protein in TAC2, which is most closely associated with airway neutrophilia and NLRP3 inflammasome activation. In a murine model of severe asthma, treatment with the MIF antagonist ISO-1 significantly inhibited neutrophilic inflammation and increased glucocorticoid responsiveness. Coimmunoprecipitation studies using lung tissue lysates demonstrated that MIF directly interacts with and cleaves annexin-A1, potentially reducing its biological activity. CONCLUSION: Our data suggest that MIF promotes glucocorticoid-resistance of neutrophilic inflammation by reducing the biological activity of annexin-A1, a potent glucocorticoid-regulated protein that inhibits neutrophil accumulation at sites of inflammation. This represents a previously unrecognised role for MIF in the regulation of inflammation and points to MIF as a potential therapeutic target for the management of severe neutrophilic asthma.


Asunto(s)
Asma , Factores Inhibidores de la Migración de Macrófagos , Humanos , Animales , Ratones , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Modelos Animales de Enfermedad , Asma/tratamiento farmacológico , Asma/metabolismo , Inflamación/metabolismo , Neutrófilos/metabolismo , Anexinas/metabolismo , Anexinas/uso terapéutico
4.
Anticancer Agents Med Chem ; 22(15): 2799-2806, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35331101

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a rampant cancer type in head and neck cancers with a poor prognosis and a high recurrence rate. Eugenol shows an anticancer effect in a variety of cancers, but it has been rarely studied in oral squamous cell carcinoma (OSCC). OBJECTIVE: The purpose of this study was to explore the role of Eugenol in OSCC and the underlying mechanism. METHODS: After different concentrations of Eugenol (0, 200, 400, and 800 µM) treatment, the viability, proliferation, migration, and invasion of OSCC cell line SCC9 were measured by CCK-8, colony formation, wound-healing, and transwell assays, respectively. TUNEL staining was employed to detect apoptosis. Western blotting was used to evaluate gene expression at the protein level. Molecular docking was used to identify the target of Eugenol. RESULTS: Eugenol decreased the proliferation and reduced the abilities of invasion and migration along with the expression of matrix metalloproteinases (MMP) 2 and MMP9 in SCC9 cells. On the contrary, the ratio of apoptotic cells was increased by Eugenol. In addition, Eugenol down-regulated B cell lymphoma-2 (Bcl-2) expression, but up-regulated BCL-2 associated X (Bax), cleaved caspase 3, and cleaved poly-ADP ribose polymerase (PARP) expression. Meanwhile, Eugenol exerted its effect on SCC9 cells in a concentration-dependent manner. Eugenol could bind to macrophage migration inhibitory factor (MIF), the expression of which was down-regulated after Eugenol treatment. Besides, overexpression of MIF reversed all the effects of Eugenol on OSCC cells. CONCLUSION: In summary, Eugenol suppressed the malignant processes of OSCC cells by targeting MIF, which could guide the clinical application of Eugenol in OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Factores Inhibidores de la Migración de Macrófagos , Neoplasias de la Boca , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Eugenol/farmacología , Eugenol/uso terapéutico , Humanos , Oxidorreductasas Intramoleculares/farmacología , Oxidorreductasas Intramoleculares/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/farmacología , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Simulación del Acoplamiento Molecular , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2 , Carcinoma de Células Escamosas de Cabeza y Cuello
5.
Oncoimmunology ; 9(1): 1846915, 2020 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-33344042

RESUMEN

Immune checkpoint blockade (ICB) has demonstrated an impressive outcome in patients with metastatic melanoma, yet, durable complete response; even with Ipilimumab/Nivolumab combo are under 30%. Primary and acquired resistance in response to ICB is commonly due to a tumor immune escape mechanism dictated by the tumor microenvironment (TME). Macrophage Migratory Inhibition Factor (MIF) has emerged as an immunosuppressive factor secreted in the TME. We have previously demonstrated that blockade of the MIF-CD74 signaling on macrophages and dendritic cells restored the anti-tumor immune response against melanoma. Here, we report that inhibition of the MIF-CD74 axis combined with ipilimumab could render resistant melanoma to better respond to anti-CTLA-4 treatment. We provide evidence that blocking the MIF-CD74 signaling potentiates CD8+ T-cells infiltration and drives pro-inflammatory M1 conversion of macrophages in the TME. Furthermore, MIF inhibition resulted in reprogramming the metabolic pathway by reducing lactate production, HIF-1α and PD-L1 expression in the resistant melanoma cells. Melanoma patient data extracted from the TCGA database supports the hypothesis that high MIF expression strongly correlates with poor response to ICB therapy. Our findings provide a rationale for combining anti-CTLA-4 with MIF inhibitors as a potential strategy to overcome resistance to ICB therapy in melanoma, turning a "cold" tumor into a "hot" one mediated by the activation of innate immunity and reprogramming of tumor metabolism and reduced PD-L1 expression in melanoma cells.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos , Melanoma , Humanos , Inhibidores de Puntos de Control Inmunológico , Oxidorreductasas Intramoleculares/uso terapéutico , Ipilimumab/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Melanoma/tratamiento farmacológico , Microambiente Tumoral
6.
Br J Clin Pharmacol ; 86(9): 1836-1848, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32207164

RESUMEN

AIM: Preclinical evidence suggests that oxidized macrophage migration inhibitory factor (oxMIF) may be involved in carcinogenesis. This phase 1 study (NCT01765790) assessed the safety, tolerability, pharmacokinetics and antitumour activity of imalumab, an oxMIF inhibitor, in patients with advanced cancer using '3 + 3' dose escalation. METHODS: In Schedule 1, patients with solid tumours received doses from 1 to 50 mg/kg IV every 2 weeks. In Schedule 2, patients with metastatic colorectal adenocarcinoma, non-small-cell lung, or ovarian cancer received weekly doses of 10 or 25 mg/kg IV (1 cycle = 28 days). Treatment continued until disease progression, unacceptable toxicity, dose-limiting toxicity, or withdrawal of consent. RESULTS: Fifty of 68 enrolled patients received imalumab. The most common treatment-related adverse events (TRAEs) included fatigue (10%) and vomiting (6%); four grade 3 serious TRAEs (two patients) occurred. The dose-limiting toxicity was allergic alveolitis (one patient, 50 mg/kg every 2 weeks). The maximum tolerated and biologically active doses were 37.5 mg/kg every 2 weeks and 10 mg/kg weekly, respectively. Of 39 assessed patients, 13 had stable disease (≥4 months in 8 patients). CONCLUSIONS: Imalumab had a maximum tolerated dose of 37.5 mg/kg every 2 weeks in patients with advanced solid tumours, with a biologically active dose of 10 mg/kg weekly. Further investigation will help define the role of oxMIF as a cancer treatment target.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Factores Inhibidores de la Migración de Macrófagos , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Masculino , Dosis Máxima Tolerada , Neoplasias/tratamiento farmacológico , Resultado del Tratamiento
7.
Parasite Immunol ; 42(4): e12698, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31976564

RESUMEN

Immunomodulatory molecules produced by helminth parasites are receiving much attention recently as novel therapeutic agents for inflammation and autoimmune diseases. In this study, we show that macrophage migration inhibitory factor (MIF) homologue from the filarial parasite, Wuchereria bancrofti (rWbaMIF-2), can suppress inflammation in a dextran sulphate sodium salt (DSS)-induced colitis model. The disease activity index (DAI) in DSS given mice showed loss of body weight and bloody diarrhoea. At autopsy, colon of these mice showed severe inflammation and reduced length. Administration of rWbaMIF-2 significantly reduced the DAI in DSS-induced colitis mice. rWbaMIF-2-treated mice had no blood in the stools, and their colon length was similar to the normal colon with minimal inflammation and histological changes. Pro-inflammatory cytokine genes (TNF-α, IL-6, IFN-γ, IL-1ß, IL-17A and NOS2) were downregulated in the colon tissue and peritoneal macrophages of rWbaMIF-2-treated mice. However, there were significant increases in IL-10-producing Treg and B1 cells in the colon and peritoneal cavity of rWbaMIF-2-treated mice. These findings suggested that rWbaMIF-2 treatment significantly ameliorated the clinical symptoms, inflammation and colon pathology in DSS given mice. This immunomodulatory effect of rWbaMIF-2 appeared to be by promoting the infiltration of Treg cells into the colon.


Asunto(s)
Colitis/tratamiento farmacológico , Oxidorreductasas Intramoleculares/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Wuchereria bancrofti , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Colon/inmunología , Colon/metabolismo , Sulfato de Dextran , Femenino , Inflamación/tratamiento farmacológico , Interleucina-17/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/metabolismo
8.
Int J Mol Med ; 41(2): 1127-1137, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207187

RESUMEN

Doxorubicin (DOXO), an anthracycline antibiotic, is a commonly used anticancer drug. Despite its widespread usage, the therapeutic effects of DOXO are limited by its cardiotoxicity. Mesenchymal stem cell (MSC)-based therapies have had positive outcomes in the treatment of DOXO-induced cardiac damage; however, DOXO exerts toxic effects on MSCs, decreasing the effectiveness of MSC therapy. Macrophage migration inhibitory factor (MIF) promotes MSC survival and rejuvenation, and thus is a promising candidate to protect MSCs against DOXO-induced injury. The present study revealed that DOXO induced the senescence of MSCs, resulting in decreased proliferation, viability and paracrine effects. However, pretreatment with MIF improved the proliferation rate, viability, paracrine function, telomere length and telomerase activity of MSCs. Furthermore, the results indicated that the molecular mechanism underlying the anti-senescent function of MIF involved the phosphatidylinositol 3-kinase (PI3K)-RAC-α serine/threonine-protein kinase (Akt) signaling pathway, which MIF activated. In agreement with this finding, silencing Akt was identified to abolish the anti-senescent effect of MIF. In addition, MIF decreased oxidative stress in MSCs, as revealed by the decreased production of reactive oxygen species and malondialdehyde, and the increased activity of superoxide dismutase. These results indicate that MIF can rescue MSCs from a state of DOXO-induced senescence by inhibiting oxidative stress and activating the PI3K-Akt signaling pathway. Thus, treatment with MIF may have an important therapeutic application for the rejuvenation of MSCs in patients with cancer being treated with DOXO.


Asunto(s)
Doxorrubicina/efectos adversos , Lesiones Cardíacas/prevención & control , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Animales , Cardiotoxicidad/genética , Cardiotoxicidad/patología , Cardiotoxicidad/prevención & control , Senescencia Celular/genética , Lesiones Cardíacas/inducido químicamente , Lesiones Cardíacas/genética , Humanos , Oxidorreductasas Intramoleculares/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal/genética
9.
Cancer Immunol Immunother ; 65(12): 1465-1476, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27629595

RESUMEN

Macrophage migration inhibitory factor (MIF) is known to be involved in oncogenic transformation, tumour progression, and immunosuppression and is overexpressed in many solid tumours, including paediatric rhabdomyosarcoma (RMS). We investigated the function of MIF in RMS during treatment with cytotoxic drugs. RMS cell lines were analysed by flow cytometry, immunofluorescence staining, and ELISA. We demonstrated the overexpression of MIF in RMS cells and the enhanced expression and secretion after treatment with cytotoxic agents. Migration assays of RMS cells revealed that inhibitors of MIF (ISO-1, Ant.III 4-IPP, Ant.V, sulforaphane (SF)) and blocking antibodies caused reduced migration, indicating a role for MIF in metastatic invasion. Additionally, we investigated the function of MIF in immune escape. The development of a population containing immunosuppressive myeloid-derived suppressor cells was promoted by incubation in conditioned medium of RMS cells comprising MIF and was reversed by MIF inhibitors but not by antibodies. Although most inhibitors may restore immune activity, Ant.III and 10 µM SF disturbed T cell proliferation in a CFSE assay, whereas T cell proliferation was not reduced by 3 µM SF, ISO-1 or antibodies. However, the inhibition of MIF by blocking antibodies did not increase the killing activity of allogenic PBMCs co-cultured with RMS cells. Our results reveal that MIF may be involved in an immune escape mechanism and demonstrate the involvement of MIF in immunogenic cell death during treatment with cytotoxic drugs. Targeting MIF may contribute to the restoration of immune sensitivity and the control of migration and metastatic invasion.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Rabdomiosarcoma/terapia , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Humanos , Factores Inhibidores de la Migración de Macrófagos/administración & dosificación , Factores Inhibidores de la Migración de Macrófagos/farmacología
10.
Cardiovasc Drugs Ther ; 30(5): 445-453, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27335054

RESUMEN

PURPOSE: Evidence suggests a two-pronged role of endogenous macrophage migration inhibitory factor (MIF) release in ischemia/reperfusion injury. We aimed to assess whether its exogenous administration confers cardioprotection. METHODS: Male C57/BL6 mice were randomly allocated to receive recombinant mouse MIF (rMIF) at physiological (ng/mL) concentrations in a dose-response fashion before or after a protocol of 35 min of ischemia and 2 h of reperfusion in an isolated Langendorff-perfused model with infarct size as endpoint. Isolated primary cardiomyocytes were also used for cell survival studies using rMIF at a supra-physiological concentration of 1 µg/mL. Pro-survival kinase activation was also studied using Western blot analyses. RESULTS: Exogenous MIF did not elicit a cardioprotective effect either when administered before the ischemic insult or when applied at reperfusion. rMIF did not confer protection when it was applied immediately before or after a hypoxia/reoxygenation insult in primary isolated cardiomyocytes. Consistently, hearts treated with MIF did not show a significant increase in phosphorylated Akt and ERK1/2. CONCLUSION: The exogenous administration of rMIF in a physiological concentration range both before ischemia and at reperfusion did not show cardioprotective effects. Although these results do not address the role of endogenous MIF after an ischemic insult followed by reperfusion, they may limit the potential translational value of rMIF.


Asunto(s)
Oxidorreductasas Intramoleculares/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas In Vitro , Oxidorreductasas Intramoleculares/farmacología , Precondicionamiento Isquémico Miocárdico , Factores Inhibidores de la Migración de Macrófagos/farmacología , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Perfusión , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/efectos de los fármacos
11.
Trends Cardiovasc Med ; 19(3): 76-86, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19679264

RESUMEN

In the recent years, atherogenesis has increasingly been linked to inflammatory processes in the injured vessel wall. Recruitment and arrest of monocytes, T cells, and neutrophils via the concerted actions of multiple chemokines and their chemokine receptors have been the subject of intense research and are being appreciated as key events underlying atherosclerotic lesion formation and progression. The evolutionary conserved cytokine macrophage migration inhibitory factor (MIF) exhibits prominent proinflammatory and proatherogenic functions, and the latest findings on its chemotactic and chemokine-like properties imply MIF as a crucial drug target for the treatment of inflammatory diseases. In this review, the role of MIF in atherosclerosis and injury-induced neointima formation is discussed. We place an emphasis on its proinflammatory and chemokine-like functions in the context of underlying extra- and intracellular signaling mechanisms. These findings clearly distinguish MIF from other cytokines in atherosclerosis and justify the intensive search for inhibitors targeting MIF in the treatment of inflammatory diseases, including advanced atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Factores Inhibidores de la Migración de Macrófagos/fisiología , Túnica Íntima/inmunología , Animales , Antígenos/inmunología , Quimiocinas/biosíntesis , Quimiocinas/inmunología , Humanos , Leucocitos/inmunología , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Metaloproteinasas de la Matriz/inmunología , Receptores CXCR
12.
Am J Physiol Lung Cell Mol Physiol ; 292(5): L1227-32, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17277045

RESUMEN

Although studies have shown that 17beta-estradiol (E(2)) prevents neutrophil infiltration and organ damage following trauma-hemorrhage, the mechanism by which E(2) inhibits neutrophil transmigration remains unknown. Macrophage migration inhibitory factor (MIF) is thought to play a central role in exacerbation of inflammation and is associated with lung injury. MIF regulates the inflammatory response through modulation of Toll-like receptor 4 (TLR4). Activation of TLR4 results in the release of proinflammatory cytokines and chemokines, which induce neutrophil infiltration and subsequent tissue damage. We hypothesized that E(2) mediates its salutary effects in the lung following trauma-hemorrhage via negative regulation of MIF and modulation of TLR4 and cytokine-induced chemotaxis. C3H/HeOuJ mice were subjected to trauma-hemorrhage (mean blood pressure 35 +/- 5 mmHg for approximately 90 min, then resuscitation) or sham operation. Mice received vehicle, E(2), or E(2) in combination with recombinant mouse MIF protein (rMIF). Trauma-hemorrhage increased lung MIF and TLR4 protein levels as well as lung and systemic levels of cytokines/chemokines. Treatment of animals with E(2) following trauma-hemorrhage prevented these changes. However, administration of rMIF protein with E(2) abolished the E(2)-mediated decrease in lung TLR4 levels, lung and plasma levels of IL-6, TNF-alpha, monocyte chemoattractant protein-1, and keratinocyte-derived chemokine (KC). Administration of rMIF protein also prevented E(2)-mediated reduction in neutrophil influx and tissue damage in the lungs following trauma-hemorrhage. These results suggest that the protective effects of E(2) on lung injury following trauma-hemorrhage are mediated via downregulation of lung MIF and TLR4-induced cytokine/chemokine production.


Asunto(s)
Estradiol/farmacología , Hemorragia/fisiopatología , Lesión Pulmonar , Factores Inhibidores de la Migración de Macrófagos/fisiología , Circulación Pulmonar/fisiología , Choque Hemorrágico/fisiopatología , Animales , Quimiocinas/sangre , Citocinas/sangre , Hemorragia/prevención & control , Pulmón/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Ratones , Ratones Endogámicos C3H , Peroxidasa/metabolismo , Choque Hemorrágico/prevención & control , Heridas y Lesiones
13.
Am J Pathol ; 167(6): 1519-29, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314467

RESUMEN

Migration inhibitory factor (MIF) responds to tissue damage and regulates inflammatory and immunological processes. To elucidate the function of MIF in cutaneous wound healing, we analyzed MIF knockout (KO) mice. After the excision of wounds from the dorsal skin of MIF KO and wild-type (WT) mice, healing was significantly delayed in MIF KO mice compared to WT mice. Lipopolysaccharide treatment significantly increased [(3)H]thymidine uptake in WT mouse fibroblasts compared to MIF KO mouse fibroblasts. Furthermore, there was a significant reduction in fibroblast and keratinocyte migration observed in MIF KO mice after 1-oleoyl-2-lysophosphatidic acid treatment. We subsequently examined whether MIF-impregnated gelatin slow-release microbeads could accelerate skin wound healing. Injection of more than 1.5 microg/500 microl of MIF-impregnated gelatin microbeads around a wound edge accelerated wound healing compared to a single MIF injection without the use of microbeads. MIF-impregnated gelatin microbeads also accelerated skin wound healing in C57BL/6 mice and diabetic db/db mice. Furthermore, incorporating MIF-impregnated gelatin microbeads into an artificial dermis implanted into MIF KO mice accelerated procollagen production and capillary formation. These findings suggest that MIF is crucial in accelerating cutaneous wound healing and that MIF-impregnated gelatin microbeads represent a promising treatment to facilitate skin wound healing.


Asunto(s)
Regeneración Tisular Dirigida/métodos , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Secuencia de Bases , Movimiento Celular/efectos de los fármacos , Cartilla de ADN , Replicación del ADN/efectos de los fármacos , Activación Enzimática , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Gelatina , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas/efectos de los fármacos
14.
Am J Pathol ; 167(6): 1561-74, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314470

RESUMEN

Age-associated differences in estrogen levels critically modify the cutaneous wound healing response. Using a microarray-based approach, we profiled changes in gene expression within the wounds of mice that were wild type or null for the pro-inflammatory cytokine macrophage migration inhibitory factor (MIF) in the presence or absence of estrogen. This experimental design identified more than 600 differentially expressed genes and established MIF as a key player in the wound healing process, regulating many novel repair/inflammation-associated gene targets. Moreover, MIF affected virtually all of the effects of reduced estrogen on wound repair. In humans, serum and wound levels of MIF increased with age and were strongly down-regulated by estrogen in vivo. Estrogen-regulated MIF transcription in vitro via a nuclear factor kappaB-dependent mechanism. These findings have wide-ranging implications for the many pathophysiological states in which MIF plays an important regulatory role and suggest a potential therapeutic role for MIF in modulating clinical conditions associated with age-related decline in estrogen levels.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos/fisiología , Cicatrización de Heridas/genética , Adulto , Envejecimiento , Animales , Ensayo de Inmunoadsorción Enzimática , Estrógenos/farmacología , Estrógenos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , Humanos , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Placebos , Regiones Promotoras Genéticas , Transcripción Genética , Transfección
16.
Vestn Oftalmol ; 121(4): 22-6, 2005.
Artículo en Ruso | MEDLINE | ID: mdl-16223038

RESUMEN

Comparative analysis of the results of combination treatment using superlymph and the conventional therapy in 17 and 52 patients, respectively, demonstrated that the combination therapy showed a marked clinical and immunological effect. The use of superlymph in local therapy, unlike the conventional etiopathogenetic treatment, resulted in an earlier decrease and arrest of an inflammatory reaction, in recovery of visual functions (for 2-4 day). Superlymph was found to be well tolerated and to cause no subjective discomfort or complications. Its immunomodulating effect was to normalize the count of T-theophylline-sensitive lymphocytes and, accordingly, the immunoregulation index, the serum concentrations of serum IgG, IgA, particularly IgE, and the phagocytic function of neutrophils. Superlymph exerted a pronounced effect on the systemic and local cytokine status, by decreasing the hypersecretion of cytokines, the regulators of inflammation. The clinical and immunological effects and good tolerability of superlymph permit it to recommend for its wide use in therapy of inflammatory eye diseases.


Asunto(s)
Citocinas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Uveítis/tratamiento farmacológico , Adolescente , Adulto , Anciano , Citocinas/administración & dosificación , Quimioterapia Combinada , Humanos , Inmunoglobulinas/análisis , Factores Inmunológicos/administración & dosificación , Recuento de Linfocitos , Factores Inhibidores de la Migración de Macrófagos/administración & dosificación , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Persona de Mediana Edad , Neutrófilos/inmunología , Fagocitosis , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Teofilina/farmacología , Factores de Tiempo , Resultado del Tratamiento , Uveítis/inmunología
17.
Infect Immun ; 73(10): 6488-92, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16177321

RESUMEN

Nosocomial infections in immune-suppressed patients are a widespread problem in intensive care medicine. Such patients are highly susceptible to infections because their immune defenses are impaired and, therefore, unable to adequately combat invading microorganisms. To investigate the problem of sepsis-induced immune suppression, we used a model in which mice developed sublethal peritonitis induced by cecal ligation and puncture (CLP). Two days after CLP mice were in an immune-suppressed state, as measured by impaired capacity to produce tumor necrosis factor (TNF) and enhanced susceptibility to bacterial infections. Since macrophage migration inhibitory factor (MIF) is a critical mediator of septic shock by modulation of innate immune responses, the role of MIF in sepsis-induced immune suppression was analyzed. Neutralization of endogenous MIF further enhanced susceptibility to bacterial superinfection after CLP. Conversely, treatment with recombinant human MIF before the bacterial superinfection protected the animals. MIF treatment reconstituted the impaired capacity to produce proinflammatory cytokines, such as TNF and interleukin-6. This study indicates that MIF might be able to ameliorate the sepsis-induced immune suppression by reenabling the organism to react adequately to a secondary bacterial challenge.


Asunto(s)
Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Tolerancia Inmunológica , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Sobreinfección/inmunología , Sobreinfección/prevención & control , Animales , Citocinas/metabolismo , Femenino , Inmunidad Innata/efectos de los fármacos , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/farmacología , Ratones , Choque Séptico/inmunología , Choque Séptico/metabolismo , Factores de Necrosis Tumoral/metabolismo
18.
Stroke ; 36(3): 613-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15692122

RESUMEN

BACKGROUND AND PURPOSE: Microglial activation may contribute to the pathogenesis of the brain injury in intracerebral hemorrhage (ICH). We have reported that the tripeptide macrophage/microglial inhibitory factor (MIF), Thr-Lys-Pro, inhibits microglial activation and results in functional improvement when given before the onset of hemorrhage. In this study, we investigate the protection and efficacy of treatment when MIF is administered 2 hours after collagenase injection. METHODS: ICH was induced by injecting bacterial collagenase into the caudate nucleus; 100 microL MIF (500 micromol/L) was delivered via a micro-osmotic pump. Infusion of MIF or saline (control) was initiated 2 hours after collagenase injection and continued for 24 or 72 hours. Microglial activation and macrophage infiltration were assessed by 5-d-4 and F4/80 immunofluorescence, respectively. Production of reactive oxygen species was visualized by in situ detection of ethidium. Degenerating neurons were assessed by Fluoro-Jade B staining. Neurological deficits, brain injury volumes, and brain edema were assessed at 24 and 72 hours after MIF/saline treatment. RESULTS: MIF can inhibit microglial activation and macrophage infiltration, attenuate the numbers of ethidium-positive cells compared with the saline-treated control mice, reduce the injury volume, edema, and degenerating neurons, and improve the neurological functional outcome. CONCLUSIONS: Activated microglia/macrophages are important contributors to brain injury after ICH. MIF could be a valuable neuroprotective agent for the treatment of ICH, if treatment is initiated soon after the onset of hemorrhage.


Asunto(s)
Lesiones Encefálicas/prevención & control , Hemorragia Cerebral/inducido químicamente , Fragmentos de Péptidos/uso terapéutico , Tuftsina/uso terapéutico , Animales , Núcleo Caudado/patología , Muerte Celular/efectos de los fármacos , Hemorragia Cerebral/complicaciones , Colagenasas/efectos adversos , Edema/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores Inhibidores de la Migración de Macrófagos/fisiología , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Neuronas/efectos de los fármacos , Trastornos Psicomotores/etiología , Trastornos Psicomotores/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Volumen Sistólico/efectos de los fármacos
19.
Nihon Rinsho ; 62(11): 2116-21, 2004 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-15552897

RESUMEN

The systemic manifestations of acute pancreatitis (AP) are responsible for the majority of pancreatitis-associated morbidity and mortality. Recent studies have established that severe AP is a disease with systemic inflammatory response syndrome as well as compensatory anti-inflammatory response syndrome. Based on their roles in the pathogenesis of AP, new therapies have been sought and tested, including those preventing the biological activity of two pro-inflammatory cytokines, tumor necrosis factor (TNF)-alpha and interleukin-1 (IL-1). Biological activity of TNF might be blunted by anti-TNF-alpha antibody or soluble TNF receptor, and IL-1 receptor antagonist might blunt that of IL-1. Although anti-cytokine therapies against IL-1, TNF-alpha or macrophage migration inhibitory factor showed promising results in experimental models of AP, the question remains as to whether similar antagonism during clinical pancreatitis would benefit patients with severe AP. Major considerations include the suitability of AP to cytokine antagonism in clinical settings, the possibility that such a therapy may lead to the development of immunosuppression and consequent infection, and whether a therapeutic window for such antagonism


Asunto(s)
Diseño de Fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Interleucina-1/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Enfermedad Aguda , Adalimumab , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Modelos Animales de Enfermedad , Etanercept , Humanos , Inmunoglobulina G/uso terapéutico , Infliximab , Factores Inhibidores de la Migración de Macrófagos/uso terapéutico , Pancreatitis/etiología , Pirrolidinas/uso terapéutico , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Índice de Severidad de la Enfermedad , Tiocarbamatos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...