Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 590
1.
Front Immunol ; 12: 753822, 2021.
Article En | MEDLINE | ID: mdl-34675935

Chronic psychosocial stress is a risk factor for the development of numerous disorders, of which most are associated with chronic low-grade inflammation. Given the immunosuppressive effects of glucocorticoids (GC), one underlying mechanism might be the development of stress-induced GC resistance in certain immune cell subpopulations. In line with this hypothesis, male mice exposed to the chronic subordinate colony housing (CSC, 19 days) model develop GC resistance of in vitro lipopolysaccharide (LPS)-stimulated splenocytes, splenomegaly and an increased percentage of splenic CD11b+ cells. Here male C57BL/6N mice were euthanized at different days during CSC, and following 30 days of single housing after stressor termination to assess when CSC-induced splenic GC resistance starts to develop and whether this is a transient effect. Moreover, splenic CD11b, GC receptor (GR) and/or macrophage migration inhibiting factor (MIF) protein levels were quantified at respective days. While mild forms of CSC-induced GC resistance, increased splenic CD11b expression and/or splenomegaly were detectable on days 8 and 9 of CSC, more severe forms took until days 15 and 16 to develop, but normalized almost completely within 30 days following stressor termination (day 51). In contrast, splenic GR expression was decreased in CSC versus single-housed control (SHC) mice at all days assessed. While MIF expression was increased on days 15 and 16 of CSC, it was decreased in CSC versus SHC mice on day 20 despite persisting splenomegaly, increased CD11b expression and functional GC resistance. In summary, our data indicate that GC resistance and CD11b+ cell-mediated splenomegaly develop gradually and in parallel over time during CSC exposure and are transient in nature. Moreover, while we can exclude that CSC-induced reduction in splenic GR expression is sufficient to induce functional GC resistance, the role of MIF in CD11b+ cell-mediated splenomegaly and GC resistance requires further investigation.


Cortisone/pharmacology , Glucocorticoids/pharmacology , Leukocytes/physiology , Spleen/cytology , Stress, Psychological/immunology , Agonistic Behavior , Animals , Bites and Stings , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Chronic Disease , Cortisone/blood , Crowding , Drug Resistance , Housing, Animal , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred C57BL , Organ Size , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Spleen/pathology , Territoriality
2.
Int J Biol Sci ; 17(7): 1837-1850, 2021.
Article En | MEDLINE | ID: mdl-33994866

Joint capsule fibrosis caused by excessive inflammation results in post-traumatic joint contracture (PTJC). Transforming growth factor (TGF)-ß1 plays a key role in PTJC by regulating fibroblast functions, however, cytokine-induced TGF-ß1 expression in specific cell types remains poorly characterized. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in inflammation- and fibrosis-associated pathophysiology. In this study, we investigated whether MIF can facilitate TGF-ß1 production from fibroblasts and regulate joint capsule fibrosis following PTJC. Our data demonstrated that MIF and TGF-ß1 significantly increased in fibroblasts of injured rat posterior joint capsules. Treatment the lesion sites with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP) reduced TGF-ß1 production and relieved joint capsule inflammation and fibrosis. In vitro, MIF facilitated TGF-ß1 expression in primary joint capsule fibroblasts by activating mitogen-activated protein kinase (MAPK) (P38, ERK) signaling through coupling with membrane surface receptor CD74, which in turn affected fibroblast functions and promoted MIF production. Our results reveal a novel function of trauma-induced MIF in the occurrence and development of joint capsule fibrosis. Further investigation of the underlying mechanism may provide potential therapeutic targets for PTJC.


Intramolecular Oxidoreductases/genetics , Joint Capsule/metabolism , Joint Diseases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/pathology , RNA/genetics , Transforming Growth Factor beta1/genetics , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Intramolecular Oxidoreductases/biosynthesis , Joint Capsule/pathology , Joint Diseases/metabolism , Joint Diseases/pathology , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophages/metabolism , Male , RNA/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Transforming Growth Factor beta1/biosynthesis
3.
Int J Mol Sci ; 22(1)2020 Dec 24.
Article En | MEDLINE | ID: mdl-33374446

Adequate tissue engineered models are required to further understand the (patho)physiological mechanism involved in the destructive processes of cartilage and subchondral bone during rheumatoid arthritis (RA). Therefore, we developed a human in vitro 3D osteochondral tissue model (OTM), mimicking cytokine-induced cellular and matrix-related changes leading to cartilage degradation and bone destruction in order to ultimately provide a preclinical drug screening tool. To this end, the OTM was engineered by co-cultivation of mesenchymal stromal cell (MSC)-derived bone and cartilage components in a 3D environment. It was comprehensively characterized on cell, protein, and mRNA level. Stimulating the OTM with pro-inflammatory cytokines, relevant in RA (tumor necrosis factor α, interleukin-6, macrophage migration inhibitory factor), caused cell- and matrix-related changes, resulting in a significantly induced gene expression of lactate dehydrogenase A, interleukin-8 and tumor necrosis factor α in both, cartilage and bone, while the matrix metalloproteases 1 and 3 were only induced in cartilage. Finally, application of target-specific drugs prevented the induction of inflammation and matrix-degradation. Thus, we here provide evidence that our human in vitro 3D OTM mimics cytokine-induced cell- and matrix-related changes-key features of RA-and may serve as a preclinical tool for the evaluation of both new targets and potential drugs in a more translational setup.


Arthritis, Rheumatoid/metabolism , Cartilage, Articular/pathology , Cytokines/metabolism , Aged , Bone and Bones/metabolism , Calcium Phosphates/metabolism , Chondrocytes/metabolism , Female , Fibroblasts/metabolism , Humans , In Vitro Techniques , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Lactate Dehydrogenase 5/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Synovial Membrane/pathology , Tissue Engineering/methods , Translational Research, Biomedical , Tumor Necrosis Factor-alpha/biosynthesis
4.
Respir Res ; 21(1): 92, 2020 Apr 22.
Article En | MEDLINE | ID: mdl-32321512

BACKGROUND: Macrophage migration inhibitory factor (MIF) has been implicated as a protective factor in the development of bronchopulmonary dysplasia (BPD) and is known to be regulated by MicroRNA-451 (miR-451). The aim of this study was to evaluate the role of miR-451 and the MIF signaling pathway in in vitro and in vivo models of BPD. METHODS: Studies were conducted in mouse lung endothelial cells (MLECs) exposed to hyperoxia and in a newborn mouse model of hyperoxia-induced BPD. Lung and cardiac morphometry as well as vascular markers were evaluated. RESULTS: Increased expression of miR-451 was noted in MLECs exposed to hyperoxia and in lungs of BPD mice. Administration of a miR-451 inhibitor to MLECs exposed to hyperoxia was associated with increased expression of MIF and decreased expression of angiopoietin (Ang) 2. Treatment with the miR-451 inhibitor was associated with improved lung morphometry indices, significant reduction in right ventricular hypertrophy, decreased mean arterial wall thickness and improvement in vascular density in BPD mice. Western blot analysis demonstrated preservation of MIF expression in BPD animals treated with a miR-451 inhibitor and increased expression of vascular endothelial growth factor-A (VEGF-A), Ang1, Ang2 and the Ang receptor, Tie2. CONCLUSION: We demonstrated that inhibition of miR-451 is associated with mitigation of the cardio-pulmonary phenotype, preservation of MIF expression and increased expression of several vascular growth factors.


Bronchopulmonary Dysplasia/metabolism , Disease Models, Animal , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Phenotype , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Cells, Cultured , Gene Expression , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , MicroRNAs/genetics , Oligonucleotides/pharmacology , Random Allocation
5.
Methods Mol Biol ; 2080: 173-183, 2020.
Article En | MEDLINE | ID: mdl-31745880

Cell death is a vital process for maintaining tissue homeostasis and removing potentially harmful cells. Cell death can be both programmed and non-programmed and is commonly divided into two main forms, termed apoptotic and necrotic death modes. In this chapter cell death is classified into apoptosis, primary necrosis, pyroptosis, and necroptosis. This chapter outlines the measurement of these different types of cell death and the relationship of measuring MIF release in these assays.


Biological Assay , Cell Death , Macrophage Migration-Inhibitory Factors/biosynthesis , Apoptosis , Biological Assay/methods , Cell Death/genetics , Humans , Macrophage Migration-Inhibitory Factors/genetics , Necrosis , Pyroptosis
6.
Alzheimers Res Ther ; 11(1): 54, 2019 06 07.
Article En | MEDLINE | ID: mdl-31174614

BACKGROUND: Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine. Chronic inflammation induced by amyloid ß proteins (Aß) is one prominent neuropathological feature in Alzheimer's disease (AD) brain. METHODS: Elisa, Western blot, and immunohistochemical staining analysis were performed to examine the level of MIF protein in CSF and brain tissues. MTT and LDH assays were used to examine the neurotoxicity, and the Morris Water Maze test was performed to examine the cognitive function in the MIF+/-/APP23 transgenic mice. RESULTS: MIF expression was upregulated in the brain of AD patients and AD model mice. Elevated MIF concentration was detected in the cerebrospinal fluid of AD patients but not in that of the patients suffering from mild cognitive impairment and vascular dementia. Reduced MIF expression impaired learning and memory in the AD model mice. MIF expression largely associates with Aß deposits and microglia. The binding assay revealed a direct association between MIF and Aß oligomers. Neurons instead of glial cells were responsible for the secretion of MIF upon stimulation by Aß oligomers. In addition, overexpression of MIF significantly protected neuronal cells from Aß-induced cytotoxicity. CONCLUSION: Our study suggests that neuronal secretion of MIF may serve as a defense mechanism to compensate for declined cognitive function in AD, and increased MIF level could be a potential AD biomarker.


Alzheimer Disease/metabolism , Brain/metabolism , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Up-Regulation/physiology , Adult , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Animals , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Female , Humans , Intramolecular Oxidoreductases/cerebrospinal fluid , Macrophage Migration-Inhibitory Factors/cerebrospinal fluid , Male , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged
7.
Nat Rev Rheumatol ; 15(7): 427-437, 2019 07.
Article En | MEDLINE | ID: mdl-31197253

The role of macrophage migration inhibitory factor (MIF) in autoimmunity is underscored by data showing that common functional polymorphisms in MIF are associated with disease susceptibility or clinical severity. MIF can regulate glucocorticoid-mediated immunosuppression and has a prominent function in cell survival signalling. Further specific functions of MIF are now being defined in different autoimmune diseases and MIF-targeted biologic therapeutics are in early-stage clinical trials. The unique structure of MIF is also directing the development of small-molecule MIF antagonists. Together, these efforts could provide a means of selectively intervening in pathogenesis and overcoming MIF-related genetic susceptibility to many rheumatic diseases.


Autoimmune Diseases/genetics , Autoimmunity , Disease Management , Genetic Predisposition to Disease , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Polymorphism, Genetic , Precision Medicine/methods , Autoimmune Diseases/therapy , Humans , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis
8.
Trends Immunol ; 40(5): 447-462, 2019 05.
Article En | MEDLINE | ID: mdl-30962001

Produced by many cell types, macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with critical and supporting roles in many disease states and conditions. Its disease associations, myriad functions, receptors, and downstream signaling have been the subject of considerable research, yet many questions remain. Moreover, the relevance of MIF's partially functionally redundant family member, D-dopachrome tautomerase (D-DT), also remains to be further characterized. Here, we discuss recent discoveries demonstrating direct roles of MIF in supporting NLR Family Pyrin Domain-Containing 3 (NRLP3) inflammasome activation, as well as acting as a molecular chaperone for intracellular proteins. These findings may offer new clues to understanding MIF's multiple functions, and assist the development of putative MIF-targeting therapeutics for a variety of pathologies.


Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Humans , Inflammasomes/immunology , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
9.
J Musculoskelet Neuronal Interact ; 18(4): 551-556, 2018 12 01.
Article En | MEDLINE | ID: mdl-30511959

OBJECTIVE: Τo investigate the effect of NF-kB signaling pathway on the expression of MIF, TNF-α, and IL-6 in the regulation of disc degeneration. METHODS: The disc tissue was taken from 56 patients with cervical spondylosis. According to the preoperative MRI and intraoperative disc herniation, the patients were divided into two groups: degeneration group and herniation group. The control group was 34 patients with cervical trauma with no history of cervical spondylosis. According to the preoperative JOA scores of cervical spondylosis, patients were divided into three groups: mild, moderate and severe. ELISA was used to detect the expression of MIF, IL-6, and TNF-α in the cervical intervertebral disc. NF-kB mRNA expression in the intervertebral disc was detected by qRT-PCR. RESULTS: The expression levels of NF-kB mRNA, MIF, IL-6 and TNF-α in the control group were significantly higher than those in the degeneration group and the herniation group (p⟨0.05). There was a positive correlation between the expression of NF-kB mRNA, MIF, IL-6, TNF- and cervical intervertebral disc degeneration. The expression of MIF, IL-6, and TNF-α in the mild, moderate, and severe group was negatively correlated with the JOA score. CONCLUSIONS: The expressions of NF-kB, MIF, IL-6, and TNF-α in intervertebral disc tissue in patients with disc herniation were increased and related to the degree of disc herniation. It may play an important role in the pathophysiological process of disc herniation.


Interleukin-6/biosynthesis , Intervertebral Disc Degeneration/metabolism , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , NF-kappa B/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Adult , Aged , Female , Gene Expression , Humans , Interleukin-6/genetics , Intervertebral Disc Degeneration/diagnosis , Intervertebral Disc Displacement/diagnosis , Intervertebral Disc Displacement/metabolism , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Male , Middle Aged , NF-kappa B/genetics , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/genetics
10.
Eur Rev Med Pharmacol Sci ; 22(15): 4908-4916, 2018 08.
Article En | MEDLINE | ID: mdl-30070326

OBJECTIVE: Lung adenocarcinoma (LA) is considered as a highly aggressive disease with heterogeneous prognosis. The molecular mechanisms of LA progression remain elusive. Recent studies have shown that dysregulation of microRNAs (miRNAs) is prevalent in LA, playing a significant role in tumor progression. The present work aims to analyze the expression and function of miR-608 in LA. PATIENTS AND METHODS: Quantitative Real-time polymerase chain reaction (qRT­PCR) assay and Western blot were performed to detect expressions of miR-608 and migration inhibitory factor (MIF). Luciferase reporter assays were carried out to investigate the regulatory effect of miR-608 on MIF. The cell invasion and the migration capabilities were detected by transwell assay. RESULTS: QRT-PCR indicated that miR-608 expressions in LA tissues were markedly reduced than that of the normal tissues. Moreover, the expression of MIF, a potential target gene of miR-608, was inversely associated with miR-608 expression in LA. Furthermore, miR-608 overexpression could inhibit LA invasion and migration, which was reversed by MIF knockdown. CONCLUSIONS: Our study revealed the mechanisms that miR-608 suppressed LA invasion and migration by targeting MIF, suggesting that miR-608/MIF axis could be used as a potential prognostic biomarker and therapeutic target for LA.


Adenocarcinoma of Lung/metabolism , Intramolecular Oxidoreductases/biosynthesis , Lung Neoplasms/metabolism , Macrophage Migration-Inhibitory Factors/biosynthesis , MicroRNAs/biosynthesis , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/genetics , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
11.
Pathol Res Pract ; 214(8): 1192-1198, 2018 Aug.
Article En | MEDLINE | ID: mdl-30041958

Macrophage migration inhibitory factor (MIF) is considered a pro-tumour factor. However, its clinical relevance in oral squamous cell carcinoma (OSCC) remains unclear. The objective of this study was to investigate the expression of MIF and its receptor CD74 in OSCC tissues, and to study the function of MIF in OSCC cells. Tissues of 90 patients with OSCC from the School of Stomatology, China Medical University were collected, and immunohistochemical staining and quantitative reverse transcription polymerase chain reaction were performed for MIF and CD74. The possible correlations between MIF and CD74 and clinical characteristics were analysed. The Kaplan-Meier analysis was used to determine the survival rates of patients. In addition, the proliferation and invasion of OSCC cells were evaluated after transfection with siRNA against MIF. MIF and CD74 levels were significantly higher in tissues of patients with OSCC than in control tissues. Moreover, MIF levels in patients with OSCC were significantly associated with cell differentiation and TNM classification. MIF expression was closely related to CD74 expression. Kaplan-Meier analysis indicated that OSCC patients with high MIF levels showed reduced overall survival and recurrenc-free survival. Furthermore, MIF expression promoted proliferation and invasion of OSCC cells. Collectively, our results reveal that MIF expression is a significant independent prognostic factor for patients with OSCC and may be a novel prognostic marker for OSCC.


Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Mouth Neoplasms/pathology , Adult , Aged , Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Cell Proliferation/physiology , Disease-Free Survival , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/biosynthesis , Humans , Intramolecular Oxidoreductases/analysis , Kaplan-Meier Estimate , Macrophage Migration-Inhibitory Factors/analysis , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Prognosis , Proportional Hazards Models , Squamous Cell Carcinoma of Head and Neck
12.
Int J Immunogenet ; 45(4): 190-200, 2018 Aug.
Article En | MEDLINE | ID: mdl-29863307

This study aimed to investigate the genetic basis of ankylosing spondylitis (AS) and polyarthralgia (PA) conditions among Indian subjects through genotyping two immune regulatory genes CD14 (-159C>T) and MIF (-173G>C) and find their association with the expression levels of three circulating inflammatory miRNAs. This investigation may provide early genetic cause of these two forms of arthritis and more optimal biological targets to predict early therapeutic outcomes. A total of 140 patients (AS: 70 and PA: 70) and 156 controls were recruited from Indian population. CD14 and MIF genotyping was performed using ARMS-PCR. Expression level of three inflammatory miRNAs (miRNA-146a, miRNA-155 and miRNA-181) was quantified using RT-qPCR. C/T genotype of CD14 gene was found to cause 2.06-fold risk of developing AS (CI 1.06-5.98, p = .04) as compared to others and G/C genotype in MIF also shown significant variation between AS and control subjects. In PA subjects, CD14 genotypes (C/T) was found to be associated with disease susceptibility and G/C genotype of MIF gene polymorphism showed 4.71-fold risk of developing PA (CI 2.58-8.62, p = .0001). The study also revealed significant upregulation of miRNA-155 expression in AS subjects (p = .0001) with more than 1.3-fold difference between AS and PA as compared to the control subjects. miRNA-155 had strong association with AS patients with CD14 genotypes (p < .05) than PA and control subjects. This study provides better understanding of the mechanisms and disease susceptibility for MIF and CD14 genetic variants and inflammatory miRNAs networks involved in AS and PA.


Arthralgia , Intramolecular Oxidoreductases , Lipopolysaccharide Receptors , Macrophage Migration-Inhibitory Factors , MicroRNAs , Polymorphism, Genetic , Spondylitis, Ankylosing , Arthralgia/genetics , Arthralgia/metabolism , Arthralgia/pathology , Female , Humans , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/genetics , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/genetics , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophage Migration-Inhibitory Factors/genetics , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology
13.
J Affect Disord ; 229: 403-409, 2018 03 15.
Article En | MEDLINE | ID: mdl-29331700

RATIONALE: Depression is associated with coronary artery disease and increases adverse outcomes and mortality in patients with acute myocardial infarction, but the underlying pathophysiological mechanisms remain unclear. OBJECTIVE: To evaluate the effect of macrophage migration inhibitory factor (MIF) on cardiac ischemia-reperfusion (I/R) injury in mice with constant darkness-induced depression. METHODS AND RESULTS: Twenty C57BL/6 mice (8 weeks old, male) were randomly divided into 2 groups: one group was housed in a 12h light/dark cycle environment (LD) and the other in a constant darkness environment (DD). After 3 weeks, constant darkness-exposed (DD) mice displayed depression-like behavior as indicated by increased immobility in the forced swim test (FST) and lower sucrose preference rate. Western blotting revealed cardiac MIF expression was significantly lower in the DD mice than that in the LD mice. Next, 84 mice were randomly divided into 4 groups: LD sham group, LD I/R group, DD sham group, and DD I/R group. Following ischemia and reperfusion, mice in the DD I/R group had a larger infarct area and lower heart function index than mice in the LD I/R group (P < 0.05 for both). The cardiac pAMPK and pACC expression levels of the DD I/R group were also lower in the DD I/R group (P < 0.05). CONCLUSION: DD-induced depression might cause decreased expression of MIF in the heart, resulting in downregulation of MIF-AMPK signaling and a subsequent adverse outcome after a cardiac I/R injury.


Darkness , Depression/metabolism , Macrophage Migration-Inhibitory Factors/biosynthesis , Myocardial Infarction/metabolism , Reperfusion Injury/metabolism , AMP-Activated Protein Kinases/biosynthesis , Animals , Depression/complications , Depression/pathology , Male , Mice , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Reperfusion Injury/pathology
14.
J Clin Pathol ; 70(4): 308-312, 2017 Apr.
Article En | MEDLINE | ID: mdl-27612504

AIMS: To investigate the expression pattern of microRNA-451 (miR-451) in patients with colorectal carcinoma and correlate with the expression of its target gene MIF (macrophage migration inhibitory factor). METHODS: Matched cancer and non-cancer fresh frozen tissues were prospectively collected from 70 patients (35 men and 35 women) who underwent resection of colorectal adenocarcinoma. These tissues collected were extracted for miR and complementary DNA conversion. Then, miR-451 expressions in these tissues were measured by quantitative real-time PCR. The expression was correlated with clinical and pathological parameters of these patients. In addition, paraffin blocks of 10 colorectal carcinomas with lowest expression of miR-451 were used for the study of MIF protein expression by immunohistochemistry. RESULTS: miR-451 was downregulated in majority of the colorectal cancer tissues when compared with their matched normal tissues (84.3%, n=59/70). Downregulation of miR-451 correlates significantly with presence of coexisting adenoma (91.4%, p=0.025). In addition, persistence of cancer or cancer recurrence after surgery showed significant correlation with downregulation of miR-451 (80% vs 0%; p=0.028). There is no significant correlation between miR-451 expression and age, gender of the patients as well as size, grades, pathological stages, presence of lymphovascular permeation, perineural invasion and microsatellite instability status of the colorectal carcinoma (p>0.05). Majority of the cases (80%) with low expression of miR-451 showed high levels of MIF protein expression confirming the inverse relationship between miR-451 and MIF expressions. CONCLUSIONS: The results showed that miR-451 could play a role in development and progression of colorectal cancer and likely by targeting MIF.


Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , MicroRNAs/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Down-Regulation , Female , Humans , Immunohistochemistry , Male , Middle Aged , Real-Time Polymerase Chain Reaction
15.
Mol Med Rep ; 13(5): 3828-34, 2016 May.
Article En | MEDLINE | ID: mdl-26985869

The aim of the present study was to investigate the expression of c-erbB-2 and macrophage migration inhibitory factor (MIF) in endometrial cancer and to elucidate the significance of the early diagnosis and prognosis of endometrial cancer. The gene copy number of c­erbB­2 and MIF was characterized by reverse transcription quantitative polymerase chain reaction and the reactivity was assessed by immunohistochemistry in 70 patients using a polyclonal antibody, and evaluated semiquantitatively according to the percentage of cells demonstrating membranous or diffuse cytoplasmic staining. A correlation between age, tumor stage, grade, myometrial invasion and lymph node metastasis was observed. The mRNA expression of c­erbB­2 and MIF was high in endometrial carcinoma. The positive expression rate of MIF protein in normal endometrium, atypical hyperplasia and endometrial carcinoma significantly increased along with the degree of aggravation of the disease by 20 (3/15), 45 (9/20) and 70% (35/50), respectively. The positive expression of MIF and c­erbB­2 was highest in endometrial cancer and a significantly higher level of protein was observed in tumors at stage I, stage G1, with a depth of myometrial invasion <0.4 cm and no lymph node metastasis. The protein expression of c­erbB­2 in endometrial cancer was higher in tumors at the G2­3 phase, clinical stage III­IV, lymph node metastasis, and had no association with the depth of myometrial invasion and age. MIF and c­erbB­2 were correlated with the occurrence and the development of endometrial cancer, and thus can be used for the early diagnosis and prognosis of endometrial cancer. The present study laid the foundation for identifying new treatments for endometrial cancer.


Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Myometrium/metabolism , Receptor, ErbB-2/biosynthesis , Adult , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Myometrium/pathology , Neoplasm Invasiveness , Neoplasm Staging
16.
Cancer Lett ; 375(2): 245-255, 2016 Jun 01.
Article En | MEDLINE | ID: mdl-26952810

Macrophage migration inhibitory factor (MIF) is up-regulated in diverse solid tumors and acts as the critical link between immune response and tumorigenesis. In this study, we demonstrated that MIF overexpression promoted migration of breast cancer cells by elevating TLR4 expression. Further investigation evidenced that MIF induced ROS generation. MIF-induced ROS led to ERK phosphorylation, which facilitated HMGB1 release from the nucleus to the cytoplasm. MIF overexpression also induced caveolin-1 phosphorylation. Caveolin-1 phosphorylation contributed to HMGB1 secretion from the cytoplasm to the extracellular matrix. The extracellular HMGB1 activated TLR4 signaling including NF-κB phosphorylation, which was responsible for the transcription of Snail and Twist as well as MMP2 activation. Furthermore, MIF-induced caveolin-1-dependent HMGB1 secretion might control the recruitment of CD11b+ immune cells. Our data suggested that MIF affected the intrinsic properties of tumors and the host immune response in tumor microenvironment by regulating the TLR4/HMGB1 axis, leading to metastasis of breast cancer.


Breast Neoplasms/genetics , HMGB1 Protein/biosynthesis , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Toll-Like Receptor 4/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cytoplasm/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , HMGB1 Protein/genetics , Humans , Immunity, Cellular/genetics , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , NF-kappa B/biosynthesis , NF-kappa B/genetics , Neoplasm Metastasis , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Toll-Like Receptor 4/biosynthesis , Tumor Microenvironment/genetics
18.
J Clin Invest ; 126(2): 732-44, 2016 Feb.
Article En | MEDLINE | ID: mdl-26752645

The immunoregulatory cytokine macrophage migration inhibitory factor (MIF) is encoded in a functionally polymorphic locus that is linked to the susceptibility of autoimmune and infectious diseases. The MIF promoter contains a 4-nucleotide microsatellite polymorphism (-794 CATT) that repeats 5 to 8 times in the locus, with greater numbers of repeats associated with higher mRNA levels. Because there is no information about the transcriptional regulation of these common alleles, we used oligonucleotide affinity chromatography and liquid chromatography-mass spectrometry to identify nuclear proteins that interact with the -794 CATT5-8 site. An analysis of monocyte nuclear lysates revealed that the transcription factor ICBP90 (also known as UHRF1) is the major protein interacting with the MIF microsatellite. We found that ICBP90 is essential for MIF transcription from monocytes/macrophages, B and T lymphocytes, and synovial fibroblasts, and TLR-induced MIF transcription is regulated in an ICBP90- and -794 CATT5-8 length-dependent manner. Whole-genome transcription analysis of ICBP90 shRNA-treated rheumatoid synoviocytes uncovered a subset of proinflammatory and immune response genes that overlapped with those regulated by MIF shRNA. In addition, the expression levels of ICBP90 and MIF were correlated in joint synovia from patients with rheumatoid arthritis. These findings identify ICBP90 as a key regulator of MIF transcription and provide functional insight into the regulation of the polymorphic MIF locus.


Arthritis, Rheumatoid/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Genetic Loci , Genetic Predisposition to Disease , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Microsatellite Repeats , Polymorphism, Genetic , Promoter Regions, Genetic , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Jurkat Cells , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Ubiquitin-Protein Ligases
19.
BMC Cancer ; 15: 843, 2015 Nov 04.
Article En | MEDLINE | ID: mdl-26530123

BACKGROUND: Poor prognosis in gallbladder cancer is due to late presentation of the disease, lack of reliable biomarkers for early diagnosis and limited targeted therapies. Early diagnostic markers and novel therapeutic targets can significantly improve clinical management of gallbladder cancer. METHODS: Proteomic analysis of four gallbladder cancer cell lines based on the invasive property (non-invasive to highly invasive) was carried out using the isobaric tags for relative and absolute quantitation labeling-based quantitative proteomic approach. The expression of macrophage migration inhibitory factor was analysed in gallbladder adenocarcinoma tissues using immunohistochemistry. In vitro cellular assays were carried out in a panel of gallbladder cancer cell lines using MIF inhibitors, ISO-1 and 4-IPP or its specific siRNA. RESULTS: The quantitative proteomic experiment led to the identification of 3,653 proteins, among which 654 were found to be overexpressed and 387 were downregulated in the invasive cell lines (OCUG-1, NOZ and GB-d1) compared to the non-invasive cell line, TGBC24TKB. Among these, macrophage migration inhibitory factor (MIF) was observed to be highly overexpressed in two of the invasive cell lines. MIF is a pleiotropic proinflammatory cytokine that plays a causative role in multiple diseases, including cancer. MIF has been reported to play a central role in tumor cell proliferation and invasion in several cancers. Immunohistochemical labeling of tumor tissue microarrays for MIF expression revealed that it was overexpressed in 21 of 29 gallbladder adenocarcinoma cases. Silencing/inhibition of MIF using siRNA and/or MIF antagonists resulted in a significant decrease in cell viability, colony forming ability and invasive property of the gallbladder cancer cells. CONCLUSIONS: Our findings support the role of MIF in tumor aggressiveness and suggest its potential application as a therapeutic target for gallbladder cancer.


Biomarkers, Tumor/biosynthesis , Gallbladder Neoplasms/genetics , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Prognosis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Early Detection of Cancer , Gallbladder Neoplasms/diagnosis , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/metabolism , Macrophages/pathology , Neoplasm Proteins/biosynthesis , Proteomics
20.
PLoS One ; 10(9): e0137366, 2015.
Article En | MEDLINE | ID: mdl-26348853

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine and has been implicated in inflammatory diseases. However, little is known about the regulation of MIF in adipose tissue and its impact on wound healing. The aim of this study was to investigate MIF expression in inflamed adipose and determine its role in inflammatory cell recruitment and wound healing. Adipose tissue was harvested from subcutaneous adipose tissue layers of 24 healthy subjects and from adipose tissue adjacent to acutely inflamed wounds of 21 patients undergoing wound debridement. MIF protein and mRNA expression were measured by ELISA and RT-PCR. Cell-specific MIF expression was visualized by immunohistochemistry. The functional role of MIF in cell recruitment was investigated by a chemotaxis assay and by flow cytometry of labeled macrophages that were injected into Mif-/-and wildtype mice. Wound healing was evaluated by an in vitro scratch assay on human fibroblast monolayers. MIF protein levels of native adipose tissue and supernatants from acutely inflamed wounds were significantly elevated when compared to healthy controls. MIF mRNA expression was increased in acutely inflamed adipose tissue indicating the activation of MIF gene transcription in response to adipose tissue inflammation. MIF is expressed in mature adipocytes and in infiltrated macrophages. Peripheral blood mononuclear cell migration was significantly increased towards supernatants derived from inflamed adipose tissue. This effect was partially abrogated by MIF-neutralizing antibodies. Moreover, when compared to wildtype mice, Mif-/-mice showed reduced infiltration of labeled macrophages into LPS-stimulated epididymal fat pads in vivo. Finally, MIF antibodies partially neutralized the detrimental effect of MIF on fibroblast wound healing. Our results indicate that increased MIF expression and rapid activation of the MIF gene in fat tissue adjacent to acute wound healing disorders may play a role in cell recruitment to the site of inflammation and wound healing.


Adipose Tissue/metabolism , Cell Movement/genetics , Inflammation/genetics , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Adult , Aged , Animals , Female , Gene Expression Regulation , Humans , Inflammation/pathology , Intramolecular Oxidoreductases/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Middle Aged , RNA, Messenger/biosynthesis , Wound Healing/genetics
...