Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Eur J Immunol ; 53(6): e2250016, 2023 06.
Article in English | MEDLINE | ID: mdl-37061852

ABSTRACT

Macrophage migration inhibitory factor (MIF) is present in high amounts in the BALF and serum of asthmatic patients, contributing to the pathogenesis of experimental asthma induced by OVA in mice. Whether MIF contributes to the physiopathology on a more complex and relevant asthma model has not been characterized. Mif-deficient (Mif-/- ) or WT mice treated with anti-MIF antibody were challenged multiple times using house dust mite (HDM) extract by the intranasal route. HDM-challenged Mif-/- mice presented decreased airway hyperresponsiveness, lung infiltration of eosinophils, mucus hypersecretion, and subepithelial fibrosis compared to HDM-challenged WT mice. Amounts of IL-4, IL-5, and IL-13 were decreased in the lungs of Mif-/- mice upon HDM challenges, but the increase of CCL11 was preserved, compared to HDM-challenged WT mice. We also observed increased numbers of group 2 innate lymphoid cells and Th2 cells in the BALF and mediastinal LNs (mLN)-induced challenged by HDM of WT mice, but not in HDM-challenged Mif-/- mice. Anti-MIF treatment abrogated the airway infiltration of eosinophils, mucus hypersecretion, and subepithelial fibrosis in the lungs of HDM-challenged mice. In conclusion, MIF ablation prevents the pathologic hallmarks of asthma in HDM-challenged mice, reinforcing the promising target of MIF for asthma therapy.


Subject(s)
Asthma , Macrophage Migration-Inhibitory Factors , Animals , Mice , Pyroglyphidae , Macrophage Migration-Inhibitory Factors/genetics , Immunity, Innate , Lymphocytes/pathology , Lung , Inflammation/pathology , Fibrosis
2.
Microb Pathog ; 173(Pt A): 105881, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36379373

ABSTRACT

Cardiomyopathy is the most serious complication of chronic Chagas disease, caused by infection with the protozoan Trypanosoma cruzi. Exacerbated inflammation of the myocardium constitutes a major pathologic component of the disease. In the myocardial microenvironment, parasite antigens and host inflammatory mediators may aggravate tissue damage. The glycoinositolphospholipid (GIPL) from T. cruzi is an inflammation-eliciting antigen recognized by Toll-like receptor 4 (TLR4), whereas the proinflammatory cytokine macrophage migration inhibitory factor (MIF) promotes progression of chronic Chagas cardiomyopathy. We herein aimed to examine the involvement of GIPL and MIF in molecular mechanisms leading to a pathogenic inflammatory response in HL-1 cardiomyocytes and HMEC microvascular endothelial cells. Immunofluorescence analysis revealed that GIPL enhanced TLR4 expression in both cell types. We found that TLR4/GIPL interaction and MIF activity modulated the arachidonic acid pathway implicated in persistent inflammation. The combination of GIPL at 50 µg/ml and MIF at 50 ng/ml upregulated type 2 cyclooxygenase (COX-2) levels in HL-1 and HMEC cells, in a stronger way than each molecule acting independently. Moreover, increased expression of prostanoid synthases and release of prostaglandin E2 (PGE2) and thromboxane B2 (TxB2) were detected in stimulated cells. Transfection experiments in HL-1 and HMEC cells showed that COX-2 induction was transcriptionally regulated through GIPL-TLR4 engagement and NFκB signaling cascade. (GIPL + MIF)-triggered NFκB activation was markedly attenuated by treatment with 100 µM Fenofibrate, a PPAR-α ligand. Fenofibrate reduced COX-2-dependent generation of bioactive lipids in HL-1 and HMEC cells. In addition, Fenofibrate abolished (GIPL + MIF)-fostered release of NO, IL-1ß, IL-6, TNF-α, and CCL2. The combined actions of GIPL and MIF display potential for amplifying the inflammatory response in myocardium of parasite-infected hosts. Our current findings might help develop more effective measures to ameliorate cardiovascular abnormalities associated with Chagas heart disease.


Subject(s)
Chagas Disease , Fenofibrate , Macrophage Migration-Inhibitory Factors , Trypanosoma cruzi , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Toll-Like Receptor 4 , Myocytes, Cardiac/metabolism , Cyclooxygenase 2 , Endothelial Cells/metabolism , Inflammation
3.
J Mammary Gland Biol Neoplasia ; 26(4): 341-355, 2021 12.
Article in English | MEDLINE | ID: mdl-34813005

ABSTRACT

Breast cancer (BC) is the leading cause of cancer-related death in women in the world. Since tumor cells employ autophagy as a survival pathway, it has been proposed that autophagy inhibition could be beneficial for cancer treatment. There are several onging clinical trials where autophagy is being inhibited (using chloroquine, CQ or hydroxychloroquine, HCQ) along with chemotherapy with promising results. However, there is also in vitro evidence in which autophagy inhibition can induce epithelial to mesenchymal transition (EMT) in cancer cells, indicating that, at least in some cases, this strategy could be detrimental for cancer patients. In this study, we found that the genetic inhibition of autophagy primed cells for EMT by inducing a decrease in E-cadherin protein levels, while CQ treatment decreased E-cadherin levels, induced morphological changes related to EMT, increased EMT-related transcription factor (EMT-TF) expression and migration in estrogen receptor positive (ER +) BC cell lines. Importantly, CQ treatment increased intracellular reactive oxygen species (ROS) which induced the secretion of macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine related to malignancy. Both ROS production and MIF secretion were responsible for the mesenchymal morphology and increased migratory capacity induced by CQ. Our results indicate that CQ treatment increased malignancy by inducing ROS production, MIF secretion and EMT and suggest that autophagy inhibition in ER + BC patients might have detrimental effects. Our data indicates that a careful selection of patients should be performed in order to determine who will benefit the most from autophagy inhibition with available pharmacological agents for the treatment of breast cancer.


Subject(s)
Breast Neoplasms , Macrophage Migration-Inhibitory Factors , Breast Neoplasms/drug therapy , Cadherins , Cell Line , Cell Line, Tumor , Chloroquine/pharmacology , Epithelial-Mesenchymal Transition , Female , Humans , Hydroxychloroquine/pharmacology , Macrophage Migration-Inhibitory Factors/pharmacology , Reactive Oxygen Species/metabolism
4.
Cell Signal ; 86: 110075, 2021 10.
Article in English | MEDLINE | ID: mdl-34229086

ABSTRACT

Autophagy can function as a survival mechanism for cancer cells and therefore, its inhibition is currently being explored as a therapy for different cancer types. For breast cancer, triple negative breast cancer (TNBC) is the subtype most sensitive to the inhibition of autophagy; but its inhibition has also been shown to promote ROS-dependent secretion of macrophage migration inhibitory factor (MIF), a pro-tumorigenic cytokine. In this work, we explore the role of MIF in breast cancer, the mechanism by which autophagy inhibition promotes MIF secretion and its effects on neighboring cancer cell signaling and macrophage polarization. We analyzed MIF mRNA expression levels in tumors from breast cancer patients from different subtypes and found that Luminal B, HER2 and Basal subtypes, which are associated to high proliferation, displayed high MIF levels. However, MIF expression had no prognostic relevance in any breast cancer subtype. In addition, we found that autophagy inhibition in 66cl4 TNBC cells increased intracellular Reactive Oxygen Species (ROS) levels, which increased MIF expression and secretion. MIF secreted from 66cl4 TNBC cells induced the activation of MIF-regulated pathways in syngeneic cell lines, increasing Akt phosphorylation in 4T1 cells and ERK phosphorylation in 67NR cells. Regarding MIF/ chemokine receptors, higher levels of CD74 and CXCR2 were found in TNBC tumor cell lines when compared to non-tumorigenic cells and CXCR7 was elevated in the highly metastatic 4T1 cell line. Finally, secreted MIF from autophagy deficient 66cl4 cells induced macrophage polarization towards the M1 subtype. Together, our results indicate an important role for the inhibition of autophagy in the regulation of ROS-mediated MIF gene expression and secretion, with paracrine effects on cancer cell signaling and pro-inflammatory repercussions in macrophage M1 polarization. This data should be considered when considering the inhibition of autophagy as a therapy for different types of cancer.


Subject(s)
Macrophage Migration-Inhibitory Factors , Triple Negative Breast Neoplasms , Autophagy , Cell Line, Tumor , Humans , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors/metabolism , Reactive Oxygen Species/metabolism
5.
Infection ; 49(3): 457-461, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33385298

ABSTRACT

PURPOSE: The establishment of candidate genetic determinants associated with tuberculosis (TB) is a challenge, considering the divergent frequencies among populations. The objective of this study was to evaluate the association between MIF - 794 CATT 5-8 polymorphism and susceptibility to TB. METHODS: Case-control study. Patients > 18 years, with pulmonary TB were included. The control group consisted of blood donors and household contacts, not relatives, healthy and > 18 years. MIF - 794 CATT 5-8 were genotyped using sequencing of PCR and capillary electrophoresis. RESULTS: 126 patients and 119 controls were included. The genotype 5/5 was more frequent among cases (15.1%) than in controls (5.9%) (p = 0.019). Cases had more frequently the allele 5 (29.4%) as compared with controls (19.3%) (p = 0.010). Prevalence of 7/X + 8/X genotypes was not different between cases and controls (p = 0.821). There was no difference between patients with alleles 7 and 8 and those with alleles 5 and 6 (p = 0.608). CONCLUSIONS: The genotype 5/5 and the allele 5 of MIF - 794 CATT 5-8 were more frequent among TB patients than in controls.


Subject(s)
Genetic Predisposition to Disease , Macrophage Migration-Inhibitory Factors , Tuberculosis , Case-Control Studies , Gene Frequency , Genotype , Humans , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors/genetics , Microsatellite Repeats , Polymorphism, Single Nucleotide , Tuberculosis/genetics
6.
Adv Clin Exp Med ; 29(5): 597-602, 2020 May.
Article in English | MEDLINE | ID: mdl-32427427

ABSTRACT

BACKGROUND: Two polymorphisms in the macrophage migration inhibitory factor (MIF) gene have been associated with inflammatory diseases (-794 CATT5-8 and -173G>C); however, so far there are no reports of studies related to oral health. OBJECTIVES: To genotype the -794 CATT5-8 and -173G>C MIF polymorphisms in Mexican patients with apical periodontitis as a genetic risk of exacerbation. MATERIAL AND METHODS: The study involved 120 patients with apical periodontitis: 60 with a diagnosis of acute apical periodontitis (Group A) and 60 without previous episodes of exacerbation (Group B). Allelic discrimination was performed from peripheral blood DNA; the repeat polymorphism -794 CATT5-8 was genotyped with sequencing, while the -173G>C polymorphism was determined using real-time polymerase chain reaction (RT-PCR) using TaqMan probes. The associations between MIF polymorphisms, haplotypes and the risk of exacerbated apical periodontitis were assessed. RESULTS: The allele CATT7 was associated with the risk of a stage of acute inflammation (OR = 4.13; 95% CI = 1.82-9.63; p =< 0.001). Regarding the -173G >C polymorphism, a process of inflammation exacerbation was only associated with the CC genotype (OR = 4.1; 95% CI = 1.02-20.84; p = 0.045). The analysis of the haplotype showed that the combination CATT7/C increases the risk of exacerbation of apical periodontitis (OR = 3.57; 95% CI = 1.038-13.300; p = 0.021). CONCLUSIONS: The polymorphisms -794 CATT5-8 and -173G>C MIF seem to significantly influence the development of a state of exacerbated inflammation in patients with apical periodontitis.


Subject(s)
Genetic Predisposition to Disease/genetics , Macrophage Migration-Inhibitory Factors/genetics , Periapical Periodontitis/genetics , Polymorphism, Genetic , Gene Frequency , Genotype , Humans , Macrophage Migration-Inhibitory Factors/blood , Polymorphism, Single Nucleotide
7.
Cytokine ; 133: 155121, 2020 09.
Article in English | MEDLINE | ID: mdl-32417648

ABSTRACT

Human macrophage migration inhibition factor (MIF) is a protein with cytokine and chemokine properties that regulates a diverse range of physiological functions related to innate immunity and inflammation. Most research has focused on the role of MIF in different inflammatory diseases. D-dopachrome tautomerase (DDT), a different molecule with structural similarities to MIF, which shares receptors and biological functions, has recently been reported, but little is known about its roles and mechanisms. In this review, we sought to understand the similarities and differences between these molecules by summarizing what is known about their different structures, receptors and mechanisms regulating their expression and biological activities with an emphasis on immunological aspects.


Subject(s)
Immunologic Factors/immunology , Immunomodulation/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Animals , Humans
8.
Inflamm Res ; 68(6): 481-491, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30944975

ABSTRACT

OBJECTIVE AND DESIGN: Respiratory syncytial virus (RSV) is the major cause of infection in children up to 2 years old and reinfection is very common among patients. Tissue damage in the lung caused by RSV leads to an immune response and infected cells activate multiple signaling pathways and massive production of inflammatory mediators like macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine. Therefore, we sought to investigate the role of MIF during RSV infection in macrophages. METHODS: We evaluated MIF expression in BALB/c mice-derived macrophages stimulated with different concentrations of RSV by Western blot and real-time PCR. Additionally, different inhibitors of signaling pathways and ROS were used to evaluate their importance for MIF expression. Furthermore, we used a specific MIF inhibitor, ISO-1, to evaluate the role of MIF in viral clearance and in RSV-induced TNF-α, MCP-1 and IL-10 release from macrophages. RESULTS: We showed that RSV induces MIF expression dependently of ROS, 5-LOX, COX and PI3K activation. Moreover, viral replication is necessary for RSV-triggered MIF expression. Differently, p38 MAPK in only partially needed for RSV-induced MIF expression. In addition, MIF is important for the release of TNF-α, MCP-1 and IL-10 triggered by RSV in macrophages. CONCLUSIONS: In conclusion, we demonstrate that MIF is expressed during RSV infection and controls the release of pro-inflammatory cytokines from macrophages in an in vitro model.


Subject(s)
Cytokines/immunology , Macrophage Migration-Inhibitory Factors/immunology , Macrophages/immunology , Respiratory Syncytial Virus Infections/immunology , Animals , Bronchoalveolar Lavage Fluid , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/virology , Mice, Inbred BALB C , Signal Transduction , Viral Load
9.
Cell Immunol ; 335: 15-21, 2019 01.
Article in English | MEDLINE | ID: mdl-30384962

ABSTRACT

Increasing attention is given to the finding that macrophages under hypoxia are capable of controlling infection by the intracellular protozoan parasite Leishmania amazonensis. The hypoxia-inducible factor (HIF)-1α has been shown to play an essential role in this enhanced innate immune response. Our study aimed to explore the HIF-1α-dependent mechanisms leading to reduced survival of the parasites residing in macrophages under low oxygen conditions. Hypoxia triggered (P < 0.01) NADPH oxidase 2 (Nox2) expression and reactive oxygen species (ROS) production in J774 macrophages upon 24-h infection with L. amazonensis. Furthermore, increased (P < 0.01) expression levels of HIF-1α and macrophage migration inhibitory factor (MIF) were detected in the infected cells grown at 3% oxygen tension. We found that either HIF-1α silencing, Nox2 inhibition or MIF antagonism caused a significant (P < 0.05) reversal of the improved leishmanicidal activity displayed by the hypoxic phagocytes. Taken together, our current results suggest that, under conditions of limited availability of oxygen, activation of the HIF-1α/MIF axis via Nox2/ROS induction promotes killing of L. amazonensis amastigotes by macrophages. Such protective mechanism might operate in L. amazonensis-infected tissues where low oxygen levels prevail.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/immunology , Animals , Cell Hypoxia , Cell Line , Hypoxia/immunology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate , Intramolecular Oxidoreductases/physiology , Leishmania/immunology , Leishmania/physiology , Macrophage Migration-Inhibitory Factors/physiology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , NADPH Oxidase 2/metabolism , NADPH Oxidase 2/physiology , Reactive Oxygen Species/metabolism
10.
Front Immunol ; 9: 1494, 2018.
Article in English | MEDLINE | ID: mdl-29997630

ABSTRACT

Understanding the mechanisms of human immunodeficiency virus type I (HIV-1) pathogenesis would facilitate the identification of new therapeutic targets to control the infection in face of current antiretroviral therapy limitations. CD74 membrane expression is upregulated in HIV-1-infected cells and the magnitude of its modulation correlates with immune hyperactivation in HIV-infected individuals. In addition, plasma level of the CD74 activating ligand macrophage migration inhibitory factor (MIF) is increased in infected subjects. However, the role played by MIF/CD74 interaction in HIV pathogenesis remains unexplored. Here, we studied the effect of MIF/CD74 interaction on primary HIV-infected monocyte-derived macrophages (MDMs) and its implications for HIV immunopathogenesis. Confocal immunofluorescence analysis of CD74 and CD44 (the MIF signal transduction co-receptor) expression indicated that both molecules colocalized at the plasma membrane specifically in wild-type HIV-infected MDMs. Treatment of infected MDMs with MIF resulted in an MIF-dependent increase in TLR4 expression. Similarly, there was a dose-dependent increase in the production of IL-6, IL-8, TNFα, IL-1ß, and sICAM compared to the no-MIF condition, specifically from infected MDMs. Importantly, the effect observed on IL-6, IL-8, TNFα, and IL-1ß was abrogated by impeding MIF interaction with CD74. Moreover, the use of a neutralizing αMIF antibody or an MIF antagonist reverted these effects, supporting the specificity of the results. Treatment of unactivated CD4+ T-cells with MIF-treated HIV-infected MDM-derived culture supernatants led to enhanced permissiveness to HIV-1 infection. This effect was lost when CD4+ T-cells were treated with supernatants derived from infected MDMs in which CD74/MIF interaction had been blocked. Moreover, the enhanced permissiveness of unactivated CD4+ T-cells was recapitulated by exogenous addition of IL-6, IL-8, IL-1ß, and TNFα, or abrogated by neutralizing its biological activity using specific antibodies. Results obtained with BAL and NL4-3 HIV laboratory strains were reproduced using transmitted/founder primary isolates. This evidence indicated that MIF/CD74 interaction resulted in a higher production of proinflammatory cytokines from HIV-infected MDMs. This caused the generation of an inflammatory microenvironment which predisposed unactivated CD4+ T-cells to HIV-1 infection, which might contribute to viral spreading and reservoir seeding. Overall, these results support a novel role of the MIF/CD74 axis in HIV pathogenesis that deserves further investigation.

11.
Cytokine ; 99: 214-224, 2017 11.
Article in English | MEDLINE | ID: mdl-28780379

ABSTRACT

BACKGROUND: Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that plays an important role in the pathogenesis of type 2 diabetes mellitus (T2DM). Although the effect of high glucose on liver function has been described, the role of MIF in hepatic mitochondrial function during T2DM has not been studied. OBJECTIVE: We examine the influence of MIF to hepatic mitochondrial function in T2DM mouse model. METHODS: WT and Mif-/- BALB/c mice were treated with a single dose of streptozotocin (STZ). After an 8-week follow-up, serum glucose, proinflammatory cytokines, C-reactive protein (CRP), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) enzyme quantification, and liver histological analyses were performed. Liver mitochondria were extracted, and mitochondrial function was evaluated by oximetry, swelling and peroxide production. RESULTS: Following treatment with STZ, WT mice (WT/STZ) developed significant hyperglycemia and high serum levels of MIF, tumor necrosis factor (TNF)-α, interleukin-ß (IL-ß), and CRP. Liver damage enzymes ALT and AST were found at high levels. In contrast, Mif-/-STZ lacked serum MIF levels and showed smaller increases in blood glucose, less TNF-α, IL-1ß, CPR, ALT and AST, and failure to develop clinical signs of disease compared to the WT/STZ group. Mitochondria extracted from the Mif-/-STZ liver showed similar respiratory control (RC) to WT/STZ or healthy mice with glutamate/malate or succinate as substrates. The four respiratory chain complexes also had comparable activities. WT/STZ-isolated mitochondria showed low swelling with calcium compared to mitochondria from Mif-/-STZ or healthy mice. Peroxide production was comparable in all groups. CONCLUSION: These results show although high systemic levels of MIF contribute to the development of T2DM pathology, the liver mitochondria remain unaltered. Importantly, the absence of MIF reduced the pathology of T2DM, also without altering liver mitochondrial function. These support MIF as a therapeutic target for the treatment of this disease in humans.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Inflammation Mediators/metabolism , Intramolecular Oxidoreductases/metabolism , Liver/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Mitochondria/metabolism , Animals , C-Reactive Protein/metabolism , Cell Respiration , Cytochromes/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Electrodes , Hyperglycemia/complications , Hyperglycemia/pathology , Interleukin-1beta/blood , Intramolecular Oxidoreductases/deficiency , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Liver/pathology , Macrophage Migration-Inhibitory Factors/deficiency , Male , Mice, Inbred BALB C , Mitochondrial Swelling , Oxygen/metabolism , Peroxides/metabolism , Streptozocin , Transaminases/metabolism , Tumor Necrosis Factor-alpha/blood
12.
J Mol Recognit ; 30(10)2017 10.
Article in English | MEDLINE | ID: mdl-28513076

ABSTRACT

The human macrophage migration inhibitory factor 1 (Hu-MIF-1) is a protein involved in the inflammatory and immunology response to parasite infection. In the present study, the existence of Hu-MIF-1 from parasites have been explored by mining WormBase. A total of 35 helminths were found to have Hu-MIF-1 homologs, including some parasites of importance for public health. Physicochemical, structural, and biological properties of Hu-MIF-1 were compared with its orthologs in parasites showing that most of these are secretory proteins, with positive net charge and presence of the Cys-Xaa-Xaa-Cys motif that is critical for its oxidoreductase activity. The inhibitor-binding site present in Hu-MIF-1 is well conserved among parasite MIFs suggesting that Hu-MIF inhibitors may target orthologs in pathogens. The binding of Hu-MIF-1 to its cognate receptor CD74 was predicted by computer-assisted docking, and it resulted to be very similar to the predicted complexes formed by parasite MIFs and human CD74. More than 1 plausible conformation of MIFs in the extracellular loops of CD74 may be possible as demonstrated by the different predicted conformations of MIF orthologs in complex with CD74. Parasite MIFs in complex with CD74 resulted with some charged residues oriented to CD74, which was not observed in the Hu-MIF-1/CD74 complex. Our findings predict the binding mode of Hu-MIF-1 and orthologs with CD74, which can assist in the design of novel MIF inhibitors. Whether the parasite MIFs function specifically subvert host immune responses to suit the parasite is an open question that needs to be further investigated. Future research should lead to a better understanding of parasite MIF action in the parasite biology.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/chemistry , Histocompatibility Antigens Class II/chemistry , Macrophage Migration-Inhibitory Factors/chemistry , Parasites/metabolism , Sequence Homology, Amino Acid , Animals , Conserved Sequence , Humans , Models, Molecular , Phylogeny , Protein Structure, Tertiary , Protein Subunits/metabolism , Sequence Alignment , Static Electricity , Structural Homology, Protein
13.
Pathol Oncol Res ; 23(2): 235-244, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27771887

ABSTRACT

Macrophage migration inhibitory factor (MIF) emerged in recent years as an important inflammation mediator, playing a prominent role in the pathogenesis of various types of malignant neoplasm. MIF is a glycoprotein that presents a wide spectrum of biological activities and exerts a complex interaction with various cellular signaling pathways, causing imbalance of homeostasis. Experimental and clinical studies show that high levels of MIF are found in almost all types of human cancers and are implicated in seemingly all stages of development of the tumors. The production of MIF is triggered through an autocrine signal emitted by tumor cells, and stimulates the production of cytokines, chemokines, and growth as well as angiogenic factors that lead to growth of the tumor, increasing its aggressiveness and metastatic potential. MIF is produced by virtually all types of human body cells, in response to stress caused by different factors, leading to pathological conditions such as chronic inflammation and immunomodulation with suppression of immune surveillance and of immune response against tumors, angiogenesis, and carcinogenesis. In this review, we present recent advances on the biological activity of MIF, the signaling pathways with which it is involved and their role in tumorigenesis.


Subject(s)
Carcinogenesis/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neoplasms/metabolism , Animals , Carcinogenesis/immunology , Humans , Immunomodulation/immunology , Inflammation/immunology , Inflammation/metabolism , Neoplasms/immunology
14.
Hum Immunol ; 77(10): 985-989, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27426952

ABSTRACT

Lepromatous Leprosy (LL) is the most common presentation of leprosy in Mexico. LL patients are unable to activate an effective inflammatory response against Mycobacterium leprae probably due to the genetics of the host. Macrophage Migration Inhibitory Factor (MIF) is important to trigger inflammation processes. Two polymorphisms have been reported for human MIF: STR -794 CATT5-8 and SNP -173 G/C. 7-8 CATT repeats at -794 and the C allele at -173 increase the expression of MIF. We aim to determine the association between the polymorphisms in MIF gene and LL. We carried a case and controls study with 100 Mexican LL patients and 100 healthy subjects (HS). PCR was used for genotyping of STR -794 CATT5-8 polymorphism and PCR-RFLP for -173 G/C. We found that LL patients possess high -794 CATT repeats (47.1%) more often than HS (32.7%). In conclusion, a MIF polymorphism is associated with susceptibility to LL in Western Mexican population.


Subject(s)
Intramolecular Oxidoreductases/genetics , Leprosy, Lepromatous/genetics , Macrophage Migration-Inhibitory Factors/genetics , Microsatellite Repeats/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Adult , Aged , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Male , Mexico , Middle Aged
15.
Clinics ; Clinics;70(3): 169-172, 03/2015. tab
Article in English | LILACS | ID: lil-747107

ABSTRACT

BACKGROUND: To evaluate the macrophage migration inhibitory factor and E-selectin levels in patients with acute coronary syndrome. MATERIALS/METHODS: We examined the plasma migration inhibitory factor and E-selectin levels in 87 patients who presented with chest pain at our hospital. The patients were classified into two groups according to their cardiac status. Sixty-five patients had acute myocardial infarction, and 22 patients had non-cardiac chest pain (non-coronary disease). We designated the latter group of patients as the control group. The patients who presented with acute myocardial infarction were further divided into two subgroups: ST-elevated myocardial infarction (n = 30) and non-ST elevated myocardial infarction (n = 35). RESULTS: We found higher plasma migration inhibitory factor levels in both acute myocardial infarction subgroups than in the control group. However, the E-selectin levels were similar between the acute myocardial infarction and control patients. In addition, we did not find a significant difference in the plasma migration inhibitory factor levels between the ST elevated myocardial infarction and NST-elevated myocardial infarction subgroups. DISCUSSION: The circulating concentrations of migration inhibitory factor were significantly increased in acute myocardial infarction patients, whereas the soluble E-selectin levels were similar between acute myocardial infarction patients and control subjects. Our results suggest that migration inhibitory factor may play a role in the atherosclerotic process. .


Subject(s)
Animals , Female , Mice , /metabolism , Interferon-gamma/metabolism , Mammary Neoplasms, Animal/immunology , Spheroids, Cellular/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Alginates , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Movement , Chitosan , /genetics , /immunology , Glucuronic Acid , Granzymes/metabolism , Hexuronic Acids , Immunity, Cellular , Interferon-gamma/genetics , Interferon-gamma/immunology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tumor Microenvironment
16.
Endocrinol Nutr ; 62(1): 31-7, 2015 Jan.
Article in English, Spanish | MEDLINE | ID: mdl-25458401

ABSTRACT

OBJECTIVE: To measure macrophage migration inhibitory factor (MIF) concentrations in obese and non-obese women diagnosed with polycystic ovary syndrome (PCOS). METHODS: Women diagnosed with PCOS and age-matched healthy controls with regular menses and normal ovaries on ultrasound examination were selected and divided into 4 groups (group A, PCOS and obese; group B, PCOS and non-obese; group C, obese controls; and group D, non-obese controls) based on body mass index (obese >30 kg/m2 and non-obese <25 kg/m2). Luteinizing hormone, follicle-stimulating hormone, androstenedione, testosterone, sex hormone-binding globulin, serum glucose, insulin and MIF levels were measured. RESULTS: Obese and non-obese women with PCOS had higher luteinizing hormone, follicle-stimulating hormone, androstenedione, testosterone, and insulin levels as compared to the obese and non-obese control groups, respectively (P < .0001). Women with PCOS had significantly higher MIF levels (group A, 48.6 ± 9.9 mg/ml; group B, 35.2 ± 6.0 ng/ml) as compared to controls (group C, 13.5 ± 6.0 ng/ml; group D, 12.0 ± 4.3 ng/dl; P < .0001). A weak, positive and significant correlation was seen between fasting blood glucose and insulin levels in women with PCOS (P < .05). CONCLUSION: Significant differences exist in plasma MIF levels between obese and non-obese women with and without PCOS.


Subject(s)
Blood Glucose , Insulin/blood , Intramolecular Oxidoreductases/blood , Macrophage Migration-Inhibitory Factors/blood , Obesity/blood , Polycystic Ovary Syndrome/blood , Adult , Androstenedione/blood , Body Mass Index , Female , Follicle Stimulating Hormone/blood , Humans , Luteinizing Hormone/blood , Ovary/diagnostic imaging , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , Young Adult
17.
Oncol Lett ; 8(5): 2267-2275, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25289107

ABSTRACT

The overexpression of macrophage migration inhibitory factor (MIF) has been identified in a variety of tumors and the investigation of its molecular mechanisms in tumor progression is a key topic of research. The present study aimed to investigate MIF as a potential marker for disease control or recurrence, and to assess the association between serum and salivary MIF and the clinicopathological characteristics of patients with oral squamous cell carcinoma (OSCC). Serum and salivary samples were collected prior to and following the surgical treatment of 50 patients with OSCC. MIF concentrations were assessed by enzyme-linked immunosorbent assay and the adopted level of statistical significance was P<0.05. The results revealed that serum MIF concentrations were significantly reduced following tumor resection in OSCC patients. Furthermore, higher preoperative salivary MIF concentrations were observed in patients with larger tumors and in those who succumbed to the disease. In conclusion, high salivary and serological MIF concentrations were identified in patients with OSCC. Nevertheless, only serological MIF concentrations may be considered as a potential marker for the early detection of OSCC recurrence once the salivary levels, prior and following treatment, do not show any significant differences.

18.
Invest. clín ; Invest. clín;55(3): 266-277, sep. 2014.
Article in Spanish | LILACS | ID: lil-780162

ABSTRACT

En diversos estudios se ha identificado que la obesidad y principalmente el aumento de adiposidad en la región abdominal, se asocia con inflamación de grado bajo, resistencia a la insulina (RI), homeostasis alterada de la glucosa y con sus comorbilidades tales como la diabetes mellitus tipo 2 (DMT2), la hipertensión, las dislipidemias y las enfermedades cardiovasculares. El factor inhibidor de la migración de macrófagos (MIF) es una citocina proinflamatoria involucrada en enfermedades autoinmunes e inflamatorias. Sin embargo, actualmente, se sugiere que el MIF está involucrado en el proceso inflamatorio que acompaña a la obesidad, así como en el control metabólico de las complicaciones asociadas a la obesidad. Los diferentes estudios muestran de manera consistente, el aumento en los niveles séricos del MIF en personas con obesidad, diabetes tipo 2 y en los diabéticos que presentan complicaciones microvasculares (la nefropatía, la retinopatía y el síndrome de pie diabético). La relación del MIF con la regulación del metabolismo de la glucosa y la apoptosis de las células β pancreáticas, así como la asociación de algunos polimorfismos funcionales en el promotor del gen del MIF con la obesidad y la diabetes. Esta revisión resume conocimientos basados en estudios clínicos y epidemiológicos sobre el papel del MIF en la obesidad y la diabetes tipo 2.


Several studies have found that obesity and increased adiposity mainly in the abdominal region, are associated with low-grade inflammation, insulin resistance (IR), impaired glucose homeostasis and comorbidities such as type 2 diabetes mellitus (T2D) and cardiovascular disease. The macrophage migration inhibitory factor (MIF), is a proinflammatory cytokine involved in autoimmune and inflammatory diseases. However, currently it is suggested that MIF is involved in the inflammatory process associated with obesity and the metabolic control of the complications associated with obesity. Different studies show consistently, increased serum levels of MIF in subjects with obesity, type 2 diabetes and diabetics with microvascular complications (nephropathy, retinopathy and diabetic foot syndrome). The relationship of the MIF to the regulation of glucose metabolism and apoptosis of pancreatic β cells, and the association of some functional polymorphisms in the promoter of the MIF gene with obesity and diabetes.This review summarizes, the knowledge based on clinical and epidemiological studies on the role of MIF in obesity and type 2 diabetes.


Subject(s)
Humans , /etiology , Macrophage Migration-Inhibitory Factors/physiology , Obesity/etiology , /blood , Macrophage Migration-Inhibitory Factors/blood , Obesity/blood
19.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;46(9): 746-751, 19/set. 2013. graf
Article in English | LILACS | ID: lil-686569

ABSTRACT

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.


Subject(s)
Animals , Mice , Hydrogen Peroxide/pharmacology , Intramolecular Oxidoreductases/drug effects , Macrophage Migration-Inhibitory Factors/drug effects , Myocytes, Cardiac/metabolism , Oxidants/pharmacology , Protein Kinase C/metabolism , src-Family Kinases/metabolism , Angiotensin II/metabolism , Blotting, Western , Cell Line , Immunohistochemistry , Intramolecular Oxidoreductases/genetics , Microscopy, Confocal , Macrophage Migration-Inhibitory Factors/genetics , Oxidative Stress/physiology , Protein Kinase Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/physiology
20.
Int J Biol Sci ; 7(9): 1298-310, 2011.
Article in English | MEDLINE | ID: mdl-22110382

ABSTRACT

Macrophage migration inhibitory factor (MIF) has been found to be involved in host resistance to several parasitic infections. To determine the mechanisms of the MIF-dependent responses to Trypanosoma cruzi, we investigated host resistance in MIF⁻/⁻ mice (on the BALB/c background) during an intraperitoneal infection. We focused on the potential involvement of MIF in dendritic cell (DC) maturation and cytokine production. Following a challenge with 5 x 10(3)T. cruzi parasites, wild type (WT) mice developed a strong IL-12 response and adequate maturation of the draining mesenteric lymph node DCs and were resistant to infection. In contrast, similarly infected MIF⁻/⁻ mice mounted a weak IL-12 response, displayed immature DCs in the early phases of infection and rapidly succumbed to T. cruzi infection. The lack of maturation and IL-12 production by the DCs in response to total T. cruzi antigen (TcAg) was confirmed by in vitro studies. These effects were reversed following treatment with recombinant MIF. Interestingly, TcAg-stimulated bone marrow-derived DCs from both WT and MIF⁻/⁻ mice had increased ERK1/2 MAPK phosphorylation. In contrast, p38 phosphorylation was only upregulated in WT DCs. Reconstitution of MIF to MIF⁻/⁻ DCs upregulated p38 phosphorylation. The MIF-p38 pathway affected MHC-II and CD86 expression as well as IL-12 production. These findings demonstrate that the MIF-induced early DC maturation and IL-12 production mediates resistance to T. cruzi infection, probably by activating the p38 pathway.


Subject(s)
Antigens, Protozoan/metabolism , Interleukin-12/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Antigens, Protozoan/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL