Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 412
Filter
1.
ACS Sens ; 8(1): 335-343, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36530142

ABSTRACT

Macrophage migration inhibitory factor (MIF), as a cytokine, plays an important role in the pathogenesis of cancer and some other diseases, and it is also one of the potential drug targets for disease treatment. However, due to the lack of simple and effective MIF imaging detection tools, the fluctuation and distribution of MIF in living cells or at lesion sites remain difficult to track precisely and in real time. Here, we report activity-based fluorescent probes, named MIFP1-MIFP3, which are used for real-time imaging and tracking of intracellular MIF, thus establishing a relationship between the fluctuation of MIF and the change of fluorescence signal during the cancer disease process. With the excellent optical properties of two-photon probe imaging, we can easily distinguish multiple cancer cells from normal cells with the representative probe, MIFP3. Moreover, MIFP3 has also been successfully used to directly identify the pathological tissues of patients with clinical liver cancer. These potential MIF probes could provide powerful tools for further study of the physiological function of MIF and will be helpful to promote the accurate diagnosis and therapeutic evaluation of MIF-associated malignancies.


Subject(s)
Macrophage Migration-Inhibitory Factors , Humans , Macrophage Migration-Inhibitory Factors/physiology , Fluorescent Dyes
3.
Cell Mol Life Sci ; 79(1): 39, 2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34921640

ABSTRACT

Traumatic brain injury (TBI), often induced by sports, car accidents, falls, or other daily occurrences, is a primary non-genetically related risk factor for the development of subsequent neurodegeneration and neuronal cell death. However, the molecular mechanisms underlying neurodegeneration, cell death, and neurobehavioral dysfunction following TBI remain unclear. Here, we found that poly(ADP-ribose) polymerase-1 (PARP-1) was hyperactivated following TBI and its inhibition reduced TBI-induced brain injury. Macrophage migration inhibitory factor (MIF), a newly identified nuclease involved in PARP-1-dependent cell death, was translocated from the cytosol to the nucleus in cortical neurons following TBI and promoted neuronal cell death in vivo. Genetic deletion of MIF protected neurons from TBI-induced dendritic spine loss, morphological complexity degeneration, and subsequent neuronal cell death in mice. Moreover, MIF knockout reduced the brain injury volume and improved long-term animal behavioral rehabilitation. These neuroprotective effects in MIF knockout mice were reversed by the expression of wild-type MIF but not nuclease-deficient MIF mutant. In contrast, genetic deletion of MIF did not alter TBI-induced neuroinflammation. These findings reveal that MIF mediates TBI-induced neurodegeneration, neuronal cell death and neurobehavioral dysfunction through its nuclease activity, but not its pro-inflammatory role. Targeting MIF's nuclease activity may offer a novel strategy to protect neurons from TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Nerve Degeneration/metabolism , Poly (ADP-Ribose) Polymerase-1/physiology , Animals , Cell Death , Male , Mice , Mice, Knockout
4.
J Biol Chem ; 297(3): 101061, 2021 09.
Article in English | MEDLINE | ID: mdl-34384784

ABSTRACT

The macrophage migration inhibitory factor (MIF) family of cytokines contains multiple ligand-binding sites and mediates immunomodulatory processes through an undefined mechanism(s). Previously, we reported a dynamic relay connecting the MIF catalytic site to an allosteric site at its solvent channel. Despite structural and functional similarity, the MIF homolog D-dopachrome tautomerase (also called MIF-2) has low sequence identity (35%), prompting the question of whether this dynamic regulatory network is conserved. Here, we establish the structural basis of an allosteric site in MIF-2, showing with solution NMR that dynamic communication is preserved in MIF-2 despite differences in the primary sequence. X-ray crystallography and NMR detail the structural consequences of perturbing residues in this pathway, which include conformational changes surrounding the allosteric site, despite global preservation of the MIF-2 fold. Molecular simulations reveal MIF-2 to contain a comparable hydrogen bond network to that of MIF, which was previously hypothesized to influence catalytic activity by modulating the strength of allosteric coupling. Disruption of the allosteric relay by mutagenesis also attenuates MIF-2 enzymatic activity in vitro and the activation of the cluster of differentiation 74 receptor in vivo, highlighting a conserved point of control for nonoverlapping functions in the MIF superfamily.


Subject(s)
Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Allosteric Site/physiology , Amino Acid Sequence/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Binding Sites/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , Cytokines/immunology , Cytokines/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Protein Binding/genetics , Structure-Activity Relationship
5.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445689

ABSTRACT

The purpose of our study was to evaluate the role of macrophage migration inhibitory factor (MIF) in the differentiation of tendon-derived stem cells (TdSCs) under hyperglycemic conditions. In the in vivo experiment, rats were classified into diabetic (DM) and non-DM groups depending on the intraperitoneal streptozotocin (STZ) or saline injection. Twelve-week after STZ injection, the supraspinatus tendon was harvested and prepared for histological evaluation and real-time reverse transcription polymerase chain reaction for osteochondrogenic (aggrecan, BMP-2, and Sox9) and tenogenic (Egr1, Mkx, scleraxis, type 1 collagen, and Tnmd) markers. For the in vitro experiment, TdSCs were isolated from healthy rat Achilles tendons. Cultured TdSCs were treated with methylglyoxal and recombinant MIF or MIF gene knockdown to determine the effect of hyperglycemic conditions and MIF on the differentiation function of TdSCs. These conditions were classified into four groups: hyperglycemic-control group, hyperglycemic-recombinant-MIF group, hyperglycemic-knockdown-MIF group, and normal-control group. The mRNA expression of osteochondrogenic and tenogenic markers was compared among the groups. In the in vivo experiment, the mRNA expression of all osteochondrogenic and tenogenic differentiation markers in the DM group was significantly higher and lower than that in the non-DM group, respectively. Similarly, in the in vitro experiments, the expression of all osteochondrogenic and tenogenic differentiation markers was significantly upregulated and downregulated, respectively, in the hyperglycemic-control group compared to that in the normal-control group. The hyperglycemic-knockdown-MIF group demonstrated significantly decreased expression of all osteochondrogenic differentiation markers and increased expression of only some tenogenic differentiation markers compared with the hyperglycemic-control group. In contrast, the hyperglycemic-recombinant-MIF group showed significantly increased expression of all osteochondrogenic differentiation markers, but no significant difference in any tenogenic marker level, compared to the hyperglycemic-control group. These results suggest that tendon homeostasis could be affected by hyperglycemic conditions, and MIF appears to alter the differentiation of TdSCs via enhancement of the osteochondrogenic differentiation in hyperglycemic conditions. These are preliminary findings, and must be confirmed in a further study.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Stem Cells/metabolism , Tendons/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Diabetes Mellitus, Experimental/physiopathology , Gene Expression/genetics , Macrophage Migration-Inhibitory Factors/pharmacology , Macrophage Migration-Inhibitory Factors/physiology , Male , Rats , Rats, Sprague-Dawley , Streptozocin/pharmacology , Tendons/physiology
6.
High Alt Med Biol ; 22(3): 317-326, 2021 09.
Article in English | MEDLINE | ID: mdl-34314630

ABSTRACT

Shaw, Snigdha, Himashree Gidugu, Gopinath Bhaumik, Maramreddy Prasanna Kumar Reddy, Usha Panjwani, and Dishari Ghosh. Anti-Mullerian hormone and macrophage migration inhibitory factor determine the reproductive health of Ladakhi women residing at 3,500 m. High Alt Med Biol. 22:317-326, 2021. Background: Reproductive health of Ladakhi high-altitude (HA) native females was investigated for the first time in this study. Available literature suggest that, female reproductive cycle and hormonal profile varies in different HA populations due to heterogeneity. Although these studies illustrate some progress on the role of HA hypoxia, it still leaves scope for evaluation of the remaining mechanisms involved in the maintenance of reproductive health in this contemporary population. Materials and Methods: Menstrual details, phasic variations in circulatory steroid hormones, and gonadotropins along with oxytocin in sea level (SL) and HA (∼3,500 m) native females of India were assessed. Moreover, ovarian reserve marker anti-Mullerian hormone (AMH) and proinflammatory cytokine macrophage migration inhibitory factor (MIF) were measured. Results: A difference in Ladakhi women was registered compared to SL, regarding luteinizing hormone (LH) (2.6 mIU/ml vs. 4.4 mIU/ml, p < 0.05) and progesterone (P) (4.1 ng/ml vs. 9.4 ng/ml, p < 0.05) levels in their luteal phase. Reduced LH might contribute to poor development of the ovarian corpus luteum, subsequently diminish P level. Decreased AMH level in three age groups: 21-30 years (1.4 ng/ml vs. 3.2 ng/ml, p < 0.01), 31-40 years (0.6 ng/ml vs. 2.1 ng/ml, p < 0.01), and >40 years (0.4 ng/ml vs. 1.7 ng/ml, p < 0.01) of Ladakhi women were recorded than their SL counterpart. Elevated oxytocin (83.5 ng/ml vs. 76.3 ng/ml, p < 0.05) and MIF levels (70.2 ng/ml vs. 49.7 ng/ml, p < 0.01) along with low P and AMH levels delineated the reason for recorded early menopause (43.9 years), shorter reproductive span (∼29 years), and history of miscarriage in HA dwellers compared to SL. Conclusion: Therefore, the findings insinuated that the response of the reproductive system to hypoxia in Ladakhi women differs from SL women, and the adaptive response in these women might be in favor of their reproductive health.


Subject(s)
Anti-Mullerian Hormone/physiology , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Adult , Anti-Mullerian Hormone/blood , Female , Humans , Intramolecular Oxidoreductases/blood , Luteinizing Hormone , Macrophage Migration-Inhibitory Factors/blood , Reproduction , Reproductive Health , Young Adult
7.
Medicine (Baltimore) ; 99(32): e21575, 2020 Aug 07.
Article in English | MEDLINE | ID: mdl-32769903

ABSTRACT

BACKGROUND: Recent studies showed that Macrophage migration inhibitory factor (MIF) is overexpressed and closely associated with prognosis in cancer patients. The present study was systematically evaluated the prognostic significance of MIF expression in cancer patients. METHODS: PubMed, Cochrane library and Scopus were searched for eligible studies up to January 2020. Pooled hazard ratio with confidence interval (CI) was determined to assess the relationship between MIF expression and survival in cancer patients. RESULTS: A total of 8 studies comprising 847 cancer patients were included in this meta-analysis. For overall survival, the pooled hazard ratio was 2.23 (95% CI 1.67-2.99, P < .001). For disease-free survival, the pooled hazard ratio was 2.24 (95% CI 1.69-2.96, P < .001). The results suggested that high expression of MIF was significantly related to poor overall survival and disease-free survival in cancer patients. CONCLUSION: MIF expression could be a valuable prognostic factor in cancer patients.


Subject(s)
Macrophage Migration-Inhibitory Factors/analysis , Neoplasms/blood , Prognosis , Chi-Square Distribution , Gene Expression/physiology , Humans , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/blood , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/blood , Macrophage Migration-Inhibitory Factors/physiology , Neoplasms/physiopathology
8.
Cell Signal ; 73: 109672, 2020 09.
Article in English | MEDLINE | ID: mdl-32428553

ABSTRACT

Overexpression of C-X-C chemokine receptor type 4 (CXCR4) has been shown in several cancers, including non-small cell lung cancer (NSCLC) and is linked to early metastasis and worse prognosis. The crosstalk between cancer cells and tumor stroma promotes the growth and metastasis and CXCR4 signaling is a key element of this crosstalk. To test the effects of CXCR4 overexpression (CXCR4-OE), we transduced the human NSCLC cell line A549 by using a lentiviral vector. A 3D cell culture model showed generations of tumorspheres and the effects derived by the co-culturing of lung fibroblasts. Using a xenograft mouse model, we also studied the effects of CXCR4-OE in pulmonary cell engraftment and tumor burden in vivo. Our data indicate that CXCR4-OE leads to increased tumorsphere formation and epithelial-mesenchymal transition (EMT). CXCR4-OE by A549 cells resulted in a significant increase in the production of the CXCR4-ligand macrophage migration inhibitory factor (MIF) compared to those transduced with an empty vector (EV) or in which the CXCR4 expression was deleted (KO). In our in vitro system, we did not detect any production of the canonical CXCR4 ligand CXCL12. Autocrine MIF production and CXCR4 signaling are part of a self-perpetuating loop that amplifies tumor growth and EMT. Co-culture with lung fibroblasts further increased tumorsphere formation, partially driven by an increase in IL-6 production. When A549 cells were injected into murine lungs, we observed more abundant and significantly larger tumor lesions in recipients of CXCR4-OE A549 cells compared to those receiving EV or KO cells, consistent with our in vitro findings. Treatment of mice with the MIF antagonist ISO-1 resulted in significantly less tumor burden. In conclusion, our data highlight the role of the CXCR4-OE/MIF/IL-6 axis in epithelial mesenchymal crosstalk and NSCLC progression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Chemokine CXCL12/metabolism , Interleukin-6/metabolism , Intramolecular Oxidoreductases/physiology , Lung Neoplasms/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Receptors, CXCR4/physiology , A549 Cells , Animals , Cell Proliferation , Epithelial-Mesenchymal Transition , Fibroblasts , Humans , Mice , Mice, Inbred NOD
9.
Transl Res ; 216: 43-56, 2020 02.
Article in English | MEDLINE | ID: mdl-31669150

ABSTRACT

Macrophage migration inhibitory factor (MIF), a pleiotropic inflammatory cytokine, is highly expressed in patients with atrial fibrillation (AF). CD74 (major histocompatibility complex, class II invariant chain) is the main receptor for MIF. However, the role of the MIF/CD74 axis in atrial arrhythmogenesis is unclear. In this study, we investigated the effects of MIF/CD74 signaling on atrial electrophysiological characteristics and determined its underlying mechanisms. Confocal fluorescence microscopy, patch clamp, and western blot analysis were used to study calcium homeostasis, ionic currents, and calcium-related signaling in MIF-treated HL-1 atrial cardiomyocytes with or without anti-CD74 neutralized antibodies treatment. Furthermore, electrocardiographic telemetry recording and echocardiography were obtained from mice treated with MIF. Compared with controls, MIF-treated HL-1 myocytes had increased calcium transients, sarcoplasmic reticulum (SR) calcium content, Na+/Ca2+ exchanger (NCX) efflux rate, calcium leak, transient outward potassium current, and ultra-rapid delayed rectifier potassium current. Furthermore, MIF could induce expression of SR Ca2+ATPase, NCX, phosphorylation of ryanodine receptor 2 (RyR2), and activation of calcium/calmodulin kinase II (CaMKII) when compared with control cells. MIF-mediated electrical dysregulation and CaMKII-RyR2 signaling activation were attenuated through blocking of CD74. Moreover, MIF-injected mice had lesser left atrium fractional shortening, greater atrial fibrosis, and atrial ectopic beats than control (nonspecific immunoglobulin treated) or MIF combined with anti-CD74 neutralized antibody-treated mice. Consequently, our study on MIF/CD74 signaling has pointed out a new potential therapeutic intervention of AF patients with MIF elevation.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Atrial Fibrillation/physiopathology , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Signal Transduction/physiology , Animals , Antibodies, Neutralizing/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Disease Progression , Histocompatibility Antigens Class II/immunology , Homeostasis , Mice
10.
Front Immunol ; 10: 2752, 2019.
Article in English | MEDLINE | ID: mdl-31866994

ABSTRACT

The macrophage migration inhibitory factor (MIF)/CD74 signaling pathway is strongly implicated in inflammation and angiogenesis. We investigated the expression of MIF and its receptor CD74 in proliferative diabetic retinopathy (PDR) to reveal a possible role of this pathway in the pathogenesis of PDR. Levels of MIF, soluble (s)CD74, soluble intercellular adhesion molecule-1 (sICAM-1) and vascular endothelial growth factor (VEGF) were significantly increased in the vitreous from patients with PDR compared to nondiabetic control samples. We detected significant positive correlations between the levels of MIF and the levels of sICAM-1 (r = 0.43; p = 0.001) and VEGF (r = 0.7; p < 0.001). Through immunohistochemical analysis of PDR epiretinal membranes, significant positive correlations were also found between microvessel density (CD31 expression) and the numbers of blood vessels expressing MIF (r = 0.56; p = 0.045) and stromal cells expressing MIF (r = 0.79; p = 0.001) and CD74 (r = 0.59; p = 0.045). Similar to VEGF, MIF was induced in Müller cells cultured under hypoxic conditions and MIF induced phosphorylation of ERK1/2 and VEGF production in Müller cells. Intravitreal administration of MIF in normal rats induced increased retinal vascular permeability and significant upregulation of phospho-ERK1/2, NF-κB, ICAM-1 and vascular cell adhesion molecule-1 expression in the retina. MIF induced migration and proliferation of human retinal microvascular endothelial cells. These results suggest that MIF/CD74 signaling is involved in PDR angiogenesis.


Subject(s)
Diabetic Retinopathy/etiology , Inflammation/etiology , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Neovascularization, Pathologic/etiology , Adult , Aged , Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Differentiation, B-Lymphocyte/physiology , Cell Movement , Cells, Cultured , Diabetic Retinopathy/physiopathology , Female , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/physiology , Humans , Intercellular Adhesion Molecule-1/analysis , Intramolecular Oxidoreductases/analysis , Macrophage Migration-Inhibitory Factors/analysis , Male , Middle Aged , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/analysis
11.
Sci Rep ; 9(1): 15507, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664114

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that increasingly is being studied in cancers and inflammatory diseases. Though murine models have been instrumental in understanding the functional role of MIF in different pathological conditions, the information obtained from these models is biased towards a specific species. In experimental science, results obtained from multiple clinically relevant animal models always provide convincing data that might recapitulate in humans. Syrian golden hamster (Mesocricetus auratus), is a clinically relevant animal model for multiple human diseases. Hence, the major objectives of this study were to characterize the structure and function of Mesocricetus auratus MIF (MaMIF) and finally evaluate its effect on pancreatic tumor growth in vivo. Initially, the recombinant MaMIF was cloned, expressed and purified in a bacterial expression system. The MaMIF primary sequence, biochemical properties, and crystal structure analysis showed greater similarity with human MIF. The crystal structure of MaMIF illustrates that it forms a homotrimer as known in human and mouse. However, MaMIF exhibits some minor structural variations when compared to human and mouse MIF. The in vitro functional studies show that MaMIF has tautomerase activity and enhances activation and migration of hamster peripheral blood mononuclear cells (PBMCs). Interestingly, injection of MaMIF into HapT1 pancreatic tumor-bearing hamsters significantly enhanced the tumor growth and tumor-associated angiogenesis. Together, the current study shows a structural and functional similarity between the hamster and human MIF. Moreover, it has demonstrated that a high level of circulating MIF originating from non-tumor cells might also promote pancreatic tumor growth in vivo.


Subject(s)
Macrophage Migration-Inhibitory Factors/physiology , Pancreatic Neoplasms/physiopathology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/physiology , Crystallography, X-Ray , Gene Knockdown Techniques , Humans , Leukocytes, Mononuclear/cytology , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/genetics , Mesocricetus , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phylogeny , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A/metabolism
12.
FASEB J ; 33(6): 6919-6932, 2019 06.
Article in English | MEDLINE | ID: mdl-30817226

ABSTRACT

T cells expressing invariant γδ antigen receptors (γδ T cells) bridge innate and adaptive immunity and facilitate barrier responses to pathogens. Macrophage migration inhibitory factor (MIF) is an upstream mediator of host defense that up-regulates the expression of pattern recognition receptors and sustains inflammatory responses by inhibiting activation-induced apoptosis in monocytes and macrophages. Surprisingly, Mif-/- γδ T cells, when compared with wild type, were observed to produce >10-fold higher levels of the proinflammatory cytokine IL-17 after stimulation with gram-positive exotoxins. High-IL-17 expression was associated with the characteristic features of IL-17-producing γδ T (γδ17) cells, including expression of IL-23R, IL-1R1, and the transcription factors RORγt and Sox13. In the gram-positive model of shock mediated by toxic shock syndrome toxin (TSST-1), Mif-/- mice succumbed to death more quickly with increased pulmonary neutrophil accumulation and higher production of cytokines, including IL-1ß and IL-23. Mif-/- γδ T cells also produced high levels of IL-17 in response to Mycobacterium lipomannan, and depletion of γδ T cells improved survival from acutely lethal Mycobacterium infection or TSST-1 administration. These data indicate that MIF deficiency is associated with a compensatory amplification of γδ17 cell responses, with implications for innate immunity and IL-17-mediated pathology in situations such as gram-positive toxic shock or Mycobacterium infection.-Kim, H. K., Garcia, A. B., Siu, E., Tilstam, P., Das, R., Roberts, S., Leng, L., Bucala, R. Macrophage migration inhibitory factor regulates innate γδ T-cell responses via IL-17 expression.


Subject(s)
Immunity, Innate/immunology , Inflammation/immunology , Interleukin-17/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/immunology , Animals , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium bovis/immunology , Receptors, Interleukin/metabolism , Shock, Septic/chemically induced , Shock, Septic/immunology , Shock, Septic/pathology , Superantigens/administration & dosage , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
13.
Nat Rev Urol ; 16(5): 318-328, 2019 05.
Article in English | MEDLINE | ID: mdl-30914802

ABSTRACT

Genitourinary cancers encompass some of the most common solid tumours and have high rates of morbidity and mortality. Inflammation is associated with enhanced tumorigenesis, and a number of pro-inflammatory mediators, such as macrophage migration inhibitory factor (MIF), also promote tumorigenesis. Studies of the role of MIF (which largely functions via the type II transmembrane receptor CD74) in prostate, bladder and kidney cancers suggest that it is a pro-tumorigenic factor in genitourinary malignancy. Inhibiting MIF activity in cell culture and in preclinical animal models of genitourinary cancers reduces the phenotypic hallmarks of cancer, such as proliferation, angiogenesis and tumour aggressiveness, by downregulating signalling pathways such as those regulated by extracellular signal-regulated kinase (ERK), protein kinase B and p53, and MIF may also reverse immunosuppression. Progress has been made in our understanding of the role of MIF (and its family member D-dopachrome tautomerase (DDT)) in genitourinary cancers and how it can be therapeutically targeted.


Subject(s)
Carcinogenesis , Macrophage Migration-Inhibitory Factors/physiology , Urogenital Neoplasms/pathology , Urogenital Neoplasms/therapy , Humans
14.
Arterioscler Thromb Vasc Biol ; 39(4): 583-592, 2019 04.
Article in English | MEDLINE | ID: mdl-30760014

ABSTRACT

With the incidence and impact of atherosclerotic cardiovascular disease and its clinical manifestations still rising, therapeutic options that target the causal mechanisms of this disorder are highly desired. Since the CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) has demonstrated that lowering inflammation can be beneficial, focusing on mechanisms underlying inflammation, for example, leukocyte recruitment, is feasible. Being key orchestrators of leukocyte trafficking, chemokines have not lost their attractiveness as therapeutic targets, despite the difficult road to drug approval thus far. Still, innovative therapeutic approaches are being developed, paving the road towards the first chemokine-based therapeutic against inflammation. In this overview, recent developments for chemokines and for the chemokine-like factor MIF (macrophage migration inhibitory factor) will be discussed.


Subject(s)
Cardiovascular Diseases/drug therapy , Chemokines/antagonists & inhibitors , Molecular Targeted Therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Chemokines/physiology , Chemotaxis, Leukocyte , Clinical Trials as Topic , Forecasting , Humans , Inflammation/complications , Inflammation/drug therapy , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/physiology , Multicenter Studies as Topic , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/physiology , Therapies, Investigational/methods , Translational Research, Biomedical
15.
Zhonghua Bing Li Xue Za Zhi ; 48(2): 108-111, 2019 Feb 08.
Article in Chinese | MEDLINE | ID: mdl-30695861

ABSTRACT

Objective: To investigate the effect of macrophage migration inhibitory factor (MIF) on the biology of glioma U87MG and U251 cells. Methods: Silencing MIF gene expression in U87MG cells by RNA interference was monitored by Western blot. MIF low expressing U251 cells were treated at different concentrations of recombinant human MIF (rhMIF) and scratching test and flow cytometry were used to detect cell migration and apoptosis. The protein expression of bcl-2, bax, AKT, p-AKT was detected by Western blot. Results: The ability of migration and anti-apoptosis of U87MG cells silenced by siRNA decreased significantly, and the expression levels of p-AKT and anti-apoptotic protein bcl-2 also decreased; in contrast, the expression level of apoptosis protein bax increased. With increase of rhMIF treatment concentration, the expression levels of MIF protein, p-AKT and bcl-2 in U251 cells were gradually enhanced, whereas the level of apoptosis protein bax was inhibited. Conclusion: MIF promotes cell migration and inhibits apoptosis of both U87MG and U251 cells, likely through the regulation of PI3K/AKT signaling pathway.


Subject(s)
Apoptosis , Brain Neoplasms/pathology , Cell Movement , Glioma/pathology , Macrophage Migration-Inhibitory Factors/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Glioma/metabolism , Humans , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , bcl-2-Associated X Protein/metabolism
16.
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
17.
Biochem Biophys Res Commun ; 508(2): 440-444, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30502082

ABSTRACT

Hyperuricemia contributes to vascular injury and dysfunction, yet the potential mechanisms are not well understood. Uric acid (UA) has been found to stimulate macrophage migration inhibitory factor (MIF) up-regulation in renal tubules from rats subjected to UA-induced nephropathy. Given that MIF is able to induce vascular smooth muscle cell (VSMC) de-differentiation (from contractile state to a secretory state), we thus hypothesized that UA-induced vascular injury is via up-regulating of MIF in VSMCs, which enhancing vascular inflammation and VSMC transition. Within a mouse model of UA injection (500 mg/kg, twice/day, 14 days), we measured circulating and vascular MIF levels under UA stimulation at 6 h, day 1, and 14. We tested the efficacy of MIF inhibitor (10 mg/kg, twice/day, 14 days) on UA-induced vascular inflammation and remodeling. High plasma level of UA induced vascular MIF release into the plasma at acute phase. In the chronic phase, the protein level of MIF is up-regulated in the vessels. MIF inhibitor suppressed vascular inflammatory responses, repressed VSMC de-differentiation, and attenuated vascular remodeling and dysfunction following UA stimulation. Knockdown of MIF in cultured VSMCs repressed UA-induced de-differentiation. Our results provided a novel mechanism for MIF-mediated vascular injury in response to UA stimulation, and suggested that anti-MIF interventions may be of therapeutic value in hyperuricemic patients.


Subject(s)
Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Vascular Remodeling/physiology , Animals , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/physiology , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Humans , Hyperuricemia/pathology , Hyperuricemia/physiopathology , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/physiology , Male , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Uric Acid/toxicity , Vascular Remodeling/drug effects , Vasculitis/chemically induced , Vasculitis/prevention & control
18.
Inflammation ; 42(3): 874-883, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30506423

ABSTRACT

Porphyromonas gingivalis (P. gingivalis) is an important pathogen that contributes to periodontal disease and causes infections that promote the progression of atherosclerosis. Our previous studies showed that macrophage migration inhibitory factor (MIF) facilitates monocyte adhesion to endothelial cells by regulating the expression of intercellular adhesion molecule-1 (ICAM-1) in P. gingivalis-infected endothelial cells. However, the detailed pathological role of MIF has yet to be elucidated in this context. To explore the functional receptor(s) of MIF that underlie its participation in the pathogenesis of atherosclerosis, we investigated the expression of the chemokine receptors CD74 and CXCR4 in endothelial cells, both of which were shown to be involved in the adhesion of monocytes to endothelial cells pretreated with P. gingivalis. Furthermore, the formation of a MIF, CD74, and CXCR4 ligand-receptor complex was revealed by our immunofluorescence staining and coimmunoprecipitation results. By interacting with the CD74/CXCR4 receptor complex, MIF may act as a crucial regulator of monocyte-endothelial cell adhesion and promote the atherosclerotic plaque formation induced by P. gingivalis.


Subject(s)
Cell Adhesion , Endothelial Cells/virology , Intercellular Adhesion Molecule-1/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Porphyromonas gingivalis/pathogenicity , Antigens, Differentiation, B-Lymphocyte , Atherosclerosis/etiology , Atherosclerosis/pathology , Atherosclerosis/virology , Cell Line , Endothelial Cells/pathology , Histocompatibility Antigens Class II , Humans , Monocytes/cytology , Receptors, CXCR4
19.
Drug Discov Today ; 24(2): 428-439, 2019 02.
Article in English | MEDLINE | ID: mdl-30439447

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a protein that acts as a cytokine-, enzyme-, endocrine- and chaperon-like molecule. It binds to the cell-surface receptor CD74 in association with CD44, which activates the downstream signal transduction pathway. In addition, MIF acts also as a noncognate ligand for C-X-C chemokine receptor type 2 (CXCR2), type 4 (CXCR4), and type 7 (CXCR7). Recently, D-dopachrome tautomerase (D-DT), a second member of the MIF superfamily, was identified. From a pharmacological and clinical point of view, the nonredundant biological properties of MIF and D-DT anticipate potential synergisms from their simultaneous inhibition. Here, we focus on the role of MIF and D-DT in human immune-inflammatory, autoimmune, and chronic respiratory diseases, providing an update on the progress made in the identification of specific small-molecule inhibitors of these proteins.


Subject(s)
Immune System Diseases/metabolism , Inflammation/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Respiratory Tract Diseases/metabolism , Animals , Chronic Disease , Humans , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors
20.
PLoS One ; 13(11): e0206368, 2018.
Article in English | MEDLINE | ID: mdl-30383785

ABSTRACT

Multiple Myeloma (MM) is the second most common hematological malignancy with a median survival of 5-10 years. While current treatments initially cause remission, relapse almost always occurs, leading to the hypothesis that a chemotherapy-resistant cancer stem cell (CSC) remains dormant, and undergoes self-renewal and differentiation to reestablish disease. Our finding is that the mature cancer cell (CD138+, rapidly proliferating and chemosensitive) has developmental plasticity; namely, the ability to dedifferentiate back into its own chemoresistant CSC progenitor, the CD138-, quiescent pre-plasma cell. We observe multiple cycles of differentiation and dedifferentiation in the absence of niche or supportive accessory cells, suggesting that soluble cytokines secreted by the MM cells themselves are responsible for this bidirectional interconversion and that stemness and chemoresistance are dynamic characteristics that can be acquired or lost and thus may be targetable. By examining cytokine secretion of CD138- and CD138+ RPMI-8226 cells, we identified that concomitant with interconversion, Macrophage Migration Inhibitory Factor (MIF-1) is secreted. The addition of a small molecule MIF-1 inhibitor (4-IPP) or MIF-1 neutralizing antibodies to CD138+ cells accelerated dedifferentiation back into the CD138- progenitor, while addition of recombinant MIF-1 drove cells towards CD138+ differentiation. A similar increase in the CD138- population is seen when MM tumor cells isolated from primary bone marrow aspirates are cultured in the presence of 4-IPP. As the CD138+ MM cell is chemosensitive, targeting MIF-1 and/or the pathways that it regulates could be a viable way to modulate stemness and chemosensitivity, which could in turn transform the treatment of MM.


Subject(s)
Cell Plasticity , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Multiple Myeloma/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cell Plasticity/drug effects , Cell Plasticity/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intramolecular Oxidoreductases/pharmacology , Macrophage Migration-Inhibitory Factors/pharmacology , Macrophages/drug effects , Macrophages/physiology , Multiple Myeloma/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Recombinant Proteins/pharmacology , Syndecan-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...