Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Article in English | MEDLINE | ID: mdl-34403986

ABSTRACT

INTRODUCTION: Prostacyclin (PGI2) is synthetized by PGI2 synthase (PGIS) and induces vasorelaxation via activation of cyclic AMP (cAMP) generating IP-receptor. Several components of the PGI2 signaling pathway are reduced in patients with pulmonary hypertension (PH). AIM: To study the effect of 17ß-estradiol (E2) on the PGI2 signaling pathway in human pulmonary arteries (HPA) and in their smooth muscle cells (hPASMC) derived from Group-3 PH and non-PH patients. METHODS: Following E2-treatments of isolated HPA and cultured hPASMC, we measured: 6-keto-Prostaglandin F1α (PGI2 stable metabolite) by ELISA, PGIS and IP protein levels by Western blot and HPA vasorelaxations with an organ bath system. RESULTS: Incubation with E2 (24/48 h, doses ≥ 10 nM) significantly increased the expression of PGIS in hPASMC derived from both PH (65-98%) and non-PH (21-33%) patients, whereas incubation with E2 (2 h, 0.1 and 1 µM) increased 6-keto-PGF1α production in HPA from Group-3 PH patients only, and did not affect 6-keto-PGF1α production in hPASMC from either non-PH or Group-3 PH patients. Increases in IP receptor expression were observed following 10 mM E2-treatment of hPASMC from non-PH (33% after 48 h) and Group-3 PH (23% after 24 h) patient lungs. Finally, preincubation with 100 nM E2 significantly increased arachidonic acid-induced vasorelaxation of HPA from non-PH patient lungs but not of HPA from Group-3 PH patient lungs. CONCLUSION: E2-treatment may help to restore the PGI2-pathway in Group-3 PH.


Subject(s)
6-Ketoprostaglandin F1 alpha/metabolism , Cytochrome P-450 Enzyme System/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Hypertension, Pulmonary/metabolism , Intramolecular Oxidoreductases/drug effects , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Vasodilation/drug effects , Antihypertensive Agents/pharmacology , Arachidonic Acid/pharmacology , Case-Control Studies , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Humans , Hypertension, Pulmonary/physiopathology , Intramolecular Oxidoreductases/metabolism , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
2.
J Neuroinflammation ; 18(1): 130, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34116703

ABSTRACT

BACKGROUND: Astrocytes are the predominant glial cell type in the central nervous system (CNS) that can secrete various cytokines and chemokines mediating neuropathology in response to danger signals. D-dopachrome tautomerase (D-DT), a newly described cytokine and a close homolog of macrophage migration inhibitory factor (MIF) protein, has been revealed to share an overlapping function with MIF in some ways. However, its cellular distribution pattern and mediated astrocyte neuropathological function in the CNS remain unclear. METHODS: A contusion model of the rat spinal cord was established. The protein levels of D-DT and PGE2 synthesis-related proteinase were assayed by Western blot and immunohistochemistry. Primary astrocytes were stimulated by different concentrations of D-DT in the presence or absence of various inhibitors to examine relevant signal pathways. The post-injury locomotor functions were assessed using the Basso, Beattie, and Bresnahan (BBB) locomotor scale. RESULTS: D-DT was inducibly expressed within astrocytes and neurons, rather than in microglia following spinal cord contusion. D-DT was able to activate the COX2/PGE2 signal pathway of astrocytes through CD74 receptor, and the intracellular activation of mitogen-activated protein kinases (MAPKs) was involved in the regulation of D-DT action. The selective inhibitor of D-DT was efficient in attenuating D-DT-induced astrocyte production of PGE2 following spinal cord injury, which contributed to the improvement of locomotor functions. CONCLUSION: Collectively, these data reveal a novel inflammatory activator of astrocytes following spinal cord injury, which might be beneficial for the development of anti-inflammation drug in neuropathological CNS.


Subject(s)
Astrocytes/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Intramolecular Oxidoreductases/metabolism , Neuroinflammatory Diseases/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Animals, Newborn , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Culture Techniques , Disease Models, Animal , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/drug effects , Locomotion/drug effects , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction
3.
Toxicol Lett ; 319: 197-203, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31785464

ABSTRACT

The chemical warfare agent sulfur mustard (SM) affects all cells in the epidermis including melanocytes which are responsible for melanin synthesis. After exposure to SM, pigment abnormalities like hypo- and hyperpigmentation can occur. The underlying molecular pathomechanisms of SM exposure on human melanogenesis have not been elucidated so far. In our study, we investigated the effect of SM on human melanocytes and melanogenesis. Normal human epidermal melanocytes (NHEM) were used as in vitro model and they were exposed to different concentrations of SM (4.5 µM-100 µM). Melanin production was analyzed by absorption measurements at 405 nm. In addition, quantitative real-time PCR (qPCR) and Western blot experiments were performed to determine the expression of essential melanogenesis-related proteins including tyrosinase (TYR), tyrosinase-related protein (TRP) 1 and 2 and microphthalmia transcription factor (MITF). Our findings demonstrated that exposure to low SM concentrations increased melanin synthesis accompanied with an increase in protein expression. In contrast, high SM concentrations led to decreased melanin content and a downregulation in expression of all investigated melanogenesis-associated proteins. We concluded that low SM concentrations may cause hyperpigmentation while high SM concentrations decreased melanin content which may explain hypopigmented skin areas in SM exposed patients.


Subject(s)
Chemical Warfare Agents/toxicity , Melanins/biosynthesis , Mustard Gas/toxicity , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Hyperpigmentation/chemically induced , Hypopigmentation/chemically induced , Intramolecular Oxidoreductases/drug effects , Melanins/genetics , Melanocytes/drug effects , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/biosynthesis , Microphthalmia-Associated Transcription Factor/genetics , Monophenol Monooxygenase/biosynthesis , Monophenol Monooxygenase/genetics , Trypsin/biosynthesis , Trypsin/genetics
4.
Alcohol Clin Exp Res ; 43(7): 1439-1451, 2019 07.
Article in English | MEDLINE | ID: mdl-31009094

ABSTRACT

BACKGROUND: Macrophage migration inhibitory factor (MIF), a pluripotent immune regulator, is an emerging mediator in alcohol-related liver disease (ALD). MIF is associated with ALD progression through its chemokine- and cytokine-like activities. METHODS: Mechanistic studies into the role of MIF in ethanol (EtOH)-induced liver injury were performed in Mif-/- mice and in C57BL/6J mice treated with a small-molecule MIF antagonist, MIF098, after Gao-Binge (acute-on-chronic) EtOH feeding, an EtOH feeding protocol associated with hepatic neutrophilia and induction of the unfolded protein response (UPR). RESULTS: The MIF axis, for example, MIF and MIF receptors invariant polypeptide of major histocompatibility complex, class II antigen-associated (CD74), CXCR2, CXCR4, and CXCR7, was enhanced in the livers of alcoholic hepatitis (AH) patients as compared to healthy controls. Mif-/- mice were protected from hepatocellular injury after Gao-Binge feeding, independent of neutrophilia and inflammation, but were associated with the UPR. Interestingly, the UPR signature in AH patients and in mice following Gao-Binge feeding was biased toward cell death with increased expression of pro-cell death CCAAT-enhancer-binding protein homologous protein (CHOP) and decreased prosurvival GRP78. The UPR and liver injury 6 hours after binge were prevented both in Mif-/- mice and in MIF098-treated mice. However, both MIF interventions led to increased liver injury and exacerbated the hepatic UPR 9 hours after binge. Induction of upstream UPR signaling and expression of CHOP protein by thapsigargin in alpha mouse liver 12 hepatocytes were blunted by coexposure to MIF098, directly connecting MIF to UPR in hepatocytes. CONCLUSIONS: The current study revealed that, in addition to its cytokine/chemokine functions, MIF is an upstream regulator of UPR in response to EtOH feeding in mice. Importantly, both MIF and UPR can either protect or contribute to liver injury, dependent upon the stage or severity of EtOH-induced liver injury.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Intramolecular Oxidoreductases/drug effects , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/drug effects , Macrophage Migration-Inhibitory Factors/genetics , Unfolded Protein Response/drug effects , Animals , Benzoxazoles/pharmacology , Endoplasmic Reticulum Chaperone BiP , Female , Granulocyte Colony-Stimulating Factor/biosynthesis , Interleukin-3/biosynthesis , Intramolecular Oxidoreductases/antagonists & inhibitors , Liver/drug effects , Liver/pathology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/pathology , Recombinant Fusion Proteins/biosynthesis
5.
Head Neck ; 39(12): 2573-2584, 2017 12.
Article in English | MEDLINE | ID: mdl-28963807

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in systemic, autoimmune, and inflammatory diseases, such as obesity, rheumatoid arthritis, and systemic lupus erythematosus. For the 2 past decades, MIF has been reported to participate in carcinogenesis, disease prognosis, tumor cell proliferation, invasion, and tumor-induced angiogenesis in many cancers. The purpose of this article is to review published experimental and clinical data for MIF and its involvement in upper aerodigestive tract cancers. Based on the current literature, we propose a biomolecular model describing the mechanisms underlying the involvement of MIF in the initiation, progression, apoptosis, and proliferation of head and neck tumor cells. In reference to this model, potential therapeutic approaches based on the use of MIF antagonists and neutralizing antibodies are described. It is concluded that MIF is a promising target for future therapeutic strategies, both with and without chemoradiation strategies.


Subject(s)
Antineoplastic Agents/administration & dosage , Head and Neck Neoplasms/pathology , Macrophage Migration-Inhibitory Factors/genetics , Molecular Targeted Therapy , Neovascularization, Pathologic/prevention & control , Apoptosis/drug effects , Apoptosis/genetics , Biopsy, Needle , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Disease Progression , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Immunohistochemistry , Intramolecular Oxidoreductases/drug effects , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/drug effects , Neovascularization, Pathologic/drug therapy , Sensitivity and Specificity , Tumor Cells, Cultured
6.
Brain Behav Immun ; 60: 27-31, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27375005

ABSTRACT

From experiments in mice in which the prostaglandin E2 (PGE2) synthesizing enzyme mPGES-1 was genetically deleted, as well as from experiments in which PGE2 was injected directly into the brain, PGE2 has been implicated as a mediator of inflammatory induced anorexia. Here we aimed at examining which PGE2 receptor (EP1-4) that was critical for the anorexic response to peripherally injected interleukin-1ß (IL-1ß). However, deletion of neither EP receptor in mice, either globally (for EP1, EP2, and EP3) or selectively in the nervous system (EP4), had any effect on the IL-1ß induced anorexia. Because these mice were all on a C57BL/6 background, whereas previous observations demonstrating a role for induced PGE2 in IL-1ß evoked anorexia had been carried out on mice on a DBA/1 background, we examined the anorexic response to IL-1ß in mice with deletion of mPGES-1 on a C57BL/6 background and a DBA/1 background, respectively. We confirmed previous findings that mPGES-1 knock-out mice on a DBA/1 background displayed attenuated anorexia to IL-1ß; however, mice on a C57BL/6 background showed the same profound anorexia as wild type mice when carrying deletion of mPGES-1, while displaying almost normal food intake after pretreatment with a cyclooxygenase-2 inhibitor. We conclude that the involvement of induced PGE2 in IL-1ß evoked anorexia is strain dependent and we suggest that different routes that probably involve distinct prostanoids exist by which inflammatory stimuli may evoke an anorexic response and that these routes may be of different importance in different strains of mice.


Subject(s)
Anorexia/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Interleukin-1beta/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/metabolism , Intramolecular Oxidoreductases/drug effects , Intramolecular Oxidoreductases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-E Synthases/pharmacology , Up-Regulation/drug effects
7.
Lupus ; 26(1): 62-72, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27230555

ABSTRACT

OBJECTIVE: Macrophage migration inhibitory factor (MIF) is a key regulator of both atherosclerosis and systemic lupus erythematosus (SLE), yet factors leading to its overproduction remain unclear. To explore regulation of MIF in SLE, we studied effects and potential mechanisms of type I interferon (IFN) and artesunate (ART), an antimalarial agent extracted from Chinese herbs, on levels of MIF. METHODS: Serum and peripheral blood cells from SLE patients and healthy controls were measured for MIF levels by ELISA and type I IFN-inducible gene expressions by real-time PCR, respectively, and assessed for associations by Spearman correlation. ART was added to human umbilical vein endothelial cell (HUVEC) cultures with or without prior IFNα-1b stimulation and to SLE peripheral blood mononuclear cell (PBMC) cultures. Protein levels of STATs and phosphorylated (p-) STATs in HUVECs were determined by Western blotting. RESULTS: Serum MIF levels were elevated in SLE patients and positively associated with disease activity (r = 0.86, p < 0.0001), accumulated damage (r = 0.34, p < 0.05), and IFN scores in SLE PBMCs (r = 0.74, p = 0.0002). The addition of IFNα-1b promoted MIF production in a time- and dose-dependent manner in HUVEC cultures. ART could inhibit expressions of IFN-inducible genes (LY6E and ISG15) in both HUVEC and SLE PBMC cultures, and suppress MIF production and over-expression of p-STAT1, but not p-STAT3 or STAT5, induced by IFNα-1b stimulation. IFNγ-induced expression of p-STAT1 in HUVECs was not inhibited by ART. CONCLUSION: MIF could be regulated by type I IFN in SLE patients. ART counteracts the effect of IFNα to inhibit MIF production by blocking STAT1 phosphorylation and thus may have therapeutic potential for SLE-associated atherosclerosis.


Subject(s)
Artemisinins/pharmacology , Intramolecular Oxidoreductases/metabolism , Lupus Erythematosus, Systemic/physiopathology , Macrophage Migration-Inhibitory Factors/metabolism , STAT1 Transcription Factor/metabolism , Adult , Antimalarials/administration & dosage , Antimalarials/pharmacology , Artemisinins/administration & dosage , Artesunate , Blotting, Western , Case-Control Studies , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Interferon Type I/metabolism , Interferon-alpha/administration & dosage , Interferon-alpha/metabolism , Intramolecular Oxidoreductases/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/drug therapy , Macrophage Migration-Inhibitory Factors/drug effects , Male , Middle Aged , Phosphorylation , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , Time Factors , Young Adult
8.
Med Res Rev ; 36(3): 440-60, 2016 May.
Article in English | MEDLINE | ID: mdl-26777977

ABSTRACT

Strong evidence has been presented linking chronic inflammation to the onset and pathogenesis of cancer. The multifunctional pro-inflammatory protein macrophage migration inhibitory factor (MIF) occupies a central role in the inflammatory pathway and has been implicated in the tumorigenesis, angiogenesis, and metastasis of many cancer phenotypes. This review highlights the current state of the art, which presents MIF, and the second member of the MIF structural superfamily, D-DT (MIF2), as significant mediators in the inflammatory-cancer axis. Although the mechanism by which MIF asserts its biological activity has yet to be fully understood, it has become clear in recent years that for certain phenotypes of cancer, MIF represents a valid therapeutic target. Current research efforts have focused on small molecule approaches that target MIF's unique tautomerase active site and neutralization of MIF with anti-MIF antibodies. These approaches have yielded promising results in a number of preclinical murine cancer models and have helped to increase our understanding of MIF biological activity. More recently, MIF's involvement in a number of key protein-protein interactions, such as with CD74 and HSP90, has been highlighted and provides a novel platform for the development of anti-MIF chemotherapeutic strategies in the future.


Subject(s)
Intramolecular Oxidoreductases/drug effects , Macrophage Migration-Inhibitory Factors/drug effects , Neoplasms/therapy , Humans , Intramolecular Oxidoreductases/chemistry , Macrophage Migration-Inhibitory Factors/chemistry , Neoplasms/metabolism
9.
J Dermatol Sci ; 80(3): 203-11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475433

ABSTRACT

BACKGROUND: The mouse pink-eyed dilution (oculocutaneous albinism II; p/Oca2(p)) locus is known to control tyrosinase activity, melanin content, and melanosome development in melanocytes. Pink-eyed dilution castaneus (p(cas)/Oca2(p-cas)) is a novel mutant allele on mouse chromosome 7 that arose spontaneously in Indonesian wild mice, Mus musculus castaneus. Mice homozygous for Oca2(p-cas) usually exhibit pink eyes and beige-colored coat on nonagouti C57BL/6 (B6) background. Recently, a novel spontaneous mutation occurred in the progeny between this mutant and B6 mice. The eyes of this novel mutant progressively become black from pink and the coat becomes dark gray from beige with aging. OBJECTIVE: The aim of this study is to clarify whatever differences exist in melanocyte proliferation and differentiation between the ordinary (pink-eyed) and novel (black-eyed) mutant using serum-free primary culture system. METHODS: The characteristics of melanocyte proliferation and differentiation were investigated by serum-free primary culture system using melanocyte-proliferation medium (MDMD). RESULTS: The proliferation of melanoblasts in MDMD did not differ between the two mice. However, when the epidermal cell suspensions were cultured with MDMD supplemented with l-tyrosine (Tyr), the differentiation of black-eyed melanocytes was greatly induced in a concentration-dependent manner compared with pink-eyed melanocytes. Immunocytochemistry demonstrated that the expression of tyrosinase and tyrosinase-related protein-1 (Tyrp1) was greatly induced or stimulated both in pink-eyed and black-eyed melanocytes, whereas the expression of microphthalmia-associated transcription factor (Mitf) was stimulated only in black-eyed melanocytes. CONCLUSION: These results suggest that the age-related coat darkening in black-eyed mutant may be caused by the increased ability of melanocyte differentiation dependent on l-Tyr through the upregulation of tyrosinase, Tyrp1, and Mitf. This mutant mouse may be useful for animal model to clarify the mechanisms of age-related pigmentation in human skin, such as melasma and solar lentigines.


Subject(s)
Aging , Albinism, Oculocutaneous/genetics , Cell Differentiation/drug effects , Melanocytes/drug effects , RNA, Messenger/metabolism , Tyrosine/pharmacology , Animals , Cell Proliferation , Intramolecular Oxidoreductases/drug effects , Intramolecular Oxidoreductases/metabolism , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Mice , Microphthalmia-Associated Transcription Factor/drug effects , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/drug effects , Monophenol Monooxygenase/metabolism , Oxidoreductases/drug effects , Oxidoreductases/metabolism , Primary Cell Culture , Up-Regulation/drug effects
10.
J Endod ; 40(12): 2024-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25266467

ABSTRACT

INTRODUCTION: The aim of this study was to evaluate the biocompatibility of a new calcium aluminate cement (EndoBinder) in subcutaneous tissue of rats in comparison with mineral trioxide aggregate and calcium hydroxide hard-setting cement. METHODS: Polyethylene tubes (1.5 × 10 mm) containing the dental cements were implanted into dorsal subcutaneous tissue of 30 rats. After experimental periods of 7, 30, and 90 days, biopsies were performed for tissue response analysis under optical light microscope. The mRNA extraction was performed for molecular evaluation of the inflammatory process in the peri-implant tissue, which was submitted to quantitative real-time polymerase chain reaction analysis for inflammatory mediators and cytokines TNF-α, Ptges2, Il-1ß, Il-4, and Il-10. RESULTS: On the basis of the score used to grade the tissue reaction (0-3), EndoBinder (0) presented no inflammatory reaction after the 90-day period, a similar result to mineral trioxide aggregate and calcium hydroxide. The thickness of inflammatory capsules (µm) also presented significant decrease during the course of periods (P < .05). As regards expression of inflammatory mediators, Ptges2 and Il-10 were detected only at 7 and 30 days, with no statistically significant difference among the experimental groups (P > .05). CONCLUSIONS: EndoBinder induced limited inflammatory reaction. It was considered biocompatible when tested in subcutaneous tissue of rats.


Subject(s)
Aluminum Compounds/pharmacology , Biocompatible Materials/pharmacology , Calcium Compounds/pharmacology , Cytokines/drug effects , Inflammation Mediators/analysis , Root Canal Filling Materials/pharmacology , Subcutaneous Tissue/drug effects , Animals , Calcium Hydroxide/pharmacology , Drug Combinations , Interleukin-10/analysis , Interleukin-1beta/drug effects , Intramolecular Oxidoreductases/drug effects , Male , Materials Testing , Oxides/pharmacology , Prostaglandin-E Synthases , Rats , Silicates/pharmacology , Subcutaneous Tissue/immunology , Subcutaneous Tissue/pathology , Time Factors , Tumor Necrosis Factor-alpha/drug effects
11.
Braz J Med Biol Res ; 46(9): 746-51, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24036910

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)
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 , Animals , Blotting, Western , Cell Line , Immunohistochemistry , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Microscopy, Confocal , Oxidative Stress/physiology , Protein Kinase Inhibitors/pharmacology , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System/physiology
12.
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
13.
J Periodontal Res ; 48(4): 527-32, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23298274

ABSTRACT

BACKGROUND AND OBJECTIVE: Macrophage migration-inhibitory factor (MIF) plays crucial roles in the recruitment and activation of macrophages as well as in helping to kill bacteria. This study investigated the expression profile of MIF in human gingiva under different periodontal conditions and its expression patterns induced by Porphyromonas gingivalis lipopolysaccharide (LPS) in gingival epithelia. MATERIAL AND METHODS: Gingival tissue samples were collected from deep pockets and clinically healthy sites of 22 nonsmoking subjects with chronic periodontitis. The expression of MIF mRNA and protein was evaluated using real-time PCR and immunohistochemistry, respectively. The in vitro study analyzed the effects of P. gingivalis LPS on the expression of MIF in a reconstituted human gingival epithelia (RHGE) model. RESULTS: In gingival epithelia, MIF protein was diffusely expressed from the basal layer to the granular and spinous layers; whereas, in the underlying connective tissues, MIF was observed around the dilated blood vessels in the deep-pocket tissues. A significantly lower level of expression of MIF mRNA and an increased level of expression of MIF protein were found in deep-pocket tissues compared with clinically healthy tissues. Expression of MIF mRNA in the RHGE model was significantly down-regulated by P. gingivalis LPS. CONCLUSION: The present study suggests that MIF expression may be related to periodontal conditions and that its expression profile could be modulated by P. gingivalis LPS. MIF may play a role in periodontal pathogenesis.


Subject(s)
Gingiva/pathology , Intramolecular Oxidoreductases/analysis , Lipopolysaccharides/pharmacology , Macrophage Migration-Inhibitory Factors/analysis , Porphyromonas gingivalis/metabolism , Adult , Capillaries/pathology , Chronic Periodontitis/pathology , Connective Tissue/blood supply , Connective Tissue/pathology , Epithelium/drug effects , Epithelium/pathology , Escherichia coli/metabolism , Gingiva/drug effects , Humans , Intramolecular Oxidoreductases/drug effects , Macrophage Migration-Inhibitory Factors/drug effects , Middle Aged , Periodontal Pocket/pathology , Tissue Culture Techniques
14.
Arch Oral Biol ; 57(9): 1274-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22480456

ABSTRACT

OBJECTIVE: This study aimed to determine in vitro how exogenous PGE(2) affects the expression of genes in cultured osteoblasts by relative quantitation PCR. DESIGN: Cultured osteoblasts were exposed to 10(-3)M, 10(-5)M or 10(-7)M PGE(2) over 5, 10, 15 and 20 days. RESULTS: RANKL expression was higher after 5 days of exposure (p<0.05), but thereafter reduced in those treated with the two lower doses of PGE(2) (p<0.01). RANKL/OPG ratio reported in favour of OPG gene expression and alkaline phosphatase gene expression increased in osteoblasts exposed to the two lower doses of the eicosanoid after 15 days. Conversely, prostaglandin E synthase, a cytokine produced during PGE(2) synthesis, gene expression was significantly reduced at 15 and 20 days (p<0.01 and 0.05 respectively). The results from this study add to the current knowledge of the mechanisms by which PGE(2) modulates the osteoblast biology in a dose-dependent manner. CONCLUSIONS: It is proposed that PGE(2)at a low dose switch osteoblast's biology in favour of bone apposition by: first, inducing a significantly higher OPG gene expression overwhelming RANKL gene expression; second, reducing PGEs synthesis; and third, increasing ALP gene expression. An opposite effect is expected when the concentration of the eicosanoid overpass certain levels.


Subject(s)
Dinoprostone/pharmacology , Osteoblasts/drug effects , Alkaline Phosphatase/analysis , Alkaline Phosphatase/drug effects , Animals , Cell Culture Techniques , Cell Shape , Core Binding Factor Alpha 1 Subunit/analysis , Dinoprostone/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Intramolecular Oxidoreductases/drug effects , Male , Osteogenesis/drug effects , Osteoprotegerin/drug effects , Prostaglandin-E Synthases , RANK Ligand/drug effects , Rats , Rats, Wistar , Time Factors
15.
Phytother Res ; 25(5): 744-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21425378

ABSTRACT

The protective effects of plant polyphenol intake on cardiovascular morbidity and mortality are widely acknowledged. Caffeine-free chicory coffee is a rich source of plant phenolics, including caffeic acid, which inhibits in vitro platelet aggregation, and also phenylpyruvate tautomerase enzymatic activity of the proinflammatory cytokine, macrophage migration inhibitory factor (MIF). To assess whether chicory coffee consumption might confer cardiovascular benefits a clinical intervention study was performed with 27 healthy volunteers, who consumed 300 mL chicory coffee every day for 1 week. The dietary intervention produced variable effects on platelet aggregation, depending on the inducer used for the aggregation test. Whole blood and plasma viscosity were both significantly decreased, along with serum MIF levels, after 1 week of chicory coffee consumption. Moreover, significant improvements were seen in red blood cell deformability. No changes in hematocrit, fibrinogen level or red blood cell counts were detected. The full spectrum of these effects is unlikely to be attributable to a single compound present in chicory coffee, nevertheless, the phenolics, including caffeic acid, are expected to play a substantial role. In conclusion, our study offers an encouraging starting-point to delineate the antithrombotic and antiinflammatory effects of phenolic compounds found in chicory coffee.


Subject(s)
Antioxidants/pharmacology , Caffeic Acids/pharmacology , Cichorium intybus/chemistry , Plant Extracts/pharmacology , Polyphenols/pharmacology , Thrombosis/prevention & control , Blood Platelets/drug effects , Blood Viscosity/drug effects , Erythrocyte Deformability/drug effects , Erythrocytes/drug effects , Feeding Behavior , Female , Humans , Intramolecular Oxidoreductases/blood , Intramolecular Oxidoreductases/drug effects , Macrophage Migration-Inhibitory Factors/blood , Macrophage Migration-Inhibitory Factors/drug effects , Male , Plant Roots/chemistry , Platelet Aggregation/drug effects , Young Adult
16.
Planta Med ; 77(13): 1504-11, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21341175

ABSTRACT

Prostaglandin E2 (PGE2) has a central role in inflammation and both cyclooxygenase-2 (COX-2) and prostaglandin E synthases are critical enzymes in its synthesis. In inflammation, bacterial products and cytokines enhance the expression of COX-2 and inducible microsomal prostaglandin E synthase-1 (mPGES-1) which are functionally coupled to result in increased PGE2 formation in macrophages and tissue cells. In the present study, we systematically investigated the effects of 26 naturally occurring flavonoids on PGE2 production and on COX-2 and mPGES-1 expression in activated macrophages. Twelve flavonoids, i.e., flavone, luteolin-7-glucoside, kaempferol, isorhamnetin, morin, quercetin, naringenin, taxifolin, pelargonidin, daidzein, genistein, and genistin effectively inhibited lipopolysaccharide (LPS)-induced PGE2 production. Four flavonoids (flavone, isorhamnetin, daidzein, and genistein) inhibited significantly LPS-induced COX-2 expression, while mPGES-1 expression was downregulated by kaempferol and isorhamnetin. The present study characterizes the effects of flavonoids on PGE2 production and on COX-2 and mPGES-1 expression in activated macrophages. The results add to our knowledge of the anti-inflammatory actions of flavonoids and introduce kaempferol and isorhamnetin as compounds capable of downregulating the expression of mPGES-1.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Flavonoids/pharmacology , Intramolecular Oxidoreductases/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Cell Line , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/analysis , Down-Regulation/drug effects , Flavonoids/chemistry , Gene Expression Regulation/drug effects , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Prostaglandin-E Synthases
17.
Clin Cancer Res ; 16(16): 4207-16, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20682710

ABSTRACT

PURPOSE: 2-(3,4-dihydroxyphenil)-ethanol (DPE), a polyphenol present in olive oil, has been found to attenuate the growth of colon cancer cells, an effect presumably related to its anti-inflammatory activity. EXPERIMENTAL DESIGN: To further explore the effects of DPE on angiogenesis and tumor growth we investigated the in vivo efficacy of DPE in a HT-29 xenograft model and in vitro activities in colon cancer cells exposed to interleukin-1beta (IL-1beta) and prostaglandin E-2 (PGE-2). RESULTS: DPE (10 mg/kg/day for 14 days) inhibited tumor growth, reducing vessel lumina and blood perfusion to tumor, and diminished expression of hypoxia inducible factor-1alpha (HIF-1alpha), vascular endothelial growth factor (VEGF), and microsomal prostaglandin-E synthase-1 (mPGEs-1). In vitro, DPE (100 mumol/L) neither affected cell proliferation nor induced apoptosis in HT-29 and WiDr cells. DPE prevented the IL-1beta-mediated increase of mPGEs-1 expression and PGE-2 generation, as it did the silencing of HIF-1alpha. Moreover, DPE blocked mPGEs-1-dependent expression of VEGF and inhibited endothelial sprouting induced by tumor cells in a coculture system. PGE-2 triggers a feed-forward loop involving HIF-1alpha, which impinges on mPGEs-1 and VEGF expression, events prevented by DPE via extracellular signal-related kinase 1/2. The reduction of PGE-2 and VEGF levels, caused by DPE, was invariably associated with a marked decrease in HIF-1alpha expression and activity, independent of proteasome activity, indicating that the DPE effects on tumor growth and angiogenesis are dependent on the inhibition of HIF-1alpha translation. CONCLUSIONS: We show that the in vivo DPE antitumor effect is associated with anti-inflammatory and antiangiogenic activities resulting from the downregulation of the HIF-1alpha/mPGEs-1/VEGF axis.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Plant Oils/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Flavonoids/pharmacology , Gene Expression/drug effects , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Intramolecular Oxidoreductases/biosynthesis , Intramolecular Oxidoreductases/drug effects , Mice , Mice, Nude , Microsomes/drug effects , Neoplasms, Experimental/metabolism , Olive Oil , Phenols/pharmacology , Plant Oils/chemistry , Polyphenols , Prostaglandin-E Synthases , RNA, Small Interfering , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
18.
Trends Cardiovasc Med ; 20(6): 189-95, 2010 Aug.
Article in English | MEDLINE | ID: mdl-22137640

ABSTRACT

Both traditional and purpose-designed nonsteroidal anti-inflammatory drugs, selective for inhibition of cyclooxygenase (COX)-2, alleviate pain and inflammation but confer a cardiovascular hazard attributable to inhibition of COX-2-derived prostacyclin (PGI(2)). Deletion of microsomal PGE synthase-1 (mPGES-1), the dominant enzyme that converts the COX-derived intermediate product PGH(2) to PGE(2), modulates inflammatory pain in rodents. In contrast with COX-2 deletion or inhibition, PGI(2) formation is augmented in mPGES-1(-/-) mice-an effect that may confer cardiovascular benefit but may undermine the analgesic potential of inhibitors of this enzyme. This review considers the cardiovascular biology of mPGES1 and the complex challenge of developing inhibitors of this enzyme.


Subject(s)
Cardiovascular Diseases/enzymology , Intramolecular Oxidoreductases/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cardiovascular Diseases/pathology , Disease Models, Animal , Endothelium, Vascular/enzymology , Humans , Inflammation , Intramolecular Oxidoreductases/antagonists & inhibitors , Neointima/enzymology , Prostaglandin-E Synthases
19.
Cancer Lett ; 290(1): 76-86, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19751963

ABSTRACT

The molecular chaperone complex Hsp90-p23 interacts with the rate-limiting catalytic subunit of telomerase, hTERT. Although their interactions are required for proper folding of nascent hTERT as well as the assembly of active telomerase, the precise role of the chaperone proteins in regulation of nuclear localization of hTERT remains unclear. Here we demonstrate that curcumin inhibits telomerase activity in a time- and dose-dependent manner by decreasing the level of hTERT expression. Following curcumin treatment, we observed a clear accumulation of hTERT in the cytoplasmic compartment of the cell. The curcumin-induced cytoplasmic retention of hTERT could be due to failure of nuclear import, and the resulting cytoplasmic hTERT protein was rapidly ubiquitinated and degraded by the proteasome. We also report that curcumin treatment results in a substantial decrease in association of p23 and hTERT but does not affect the Hsp90 binding to hTERT. In contrast, the treatment of the Hsp90 inhibitor geldanamycin promotes dissociation of both Hsp90 and p23 proteins from hTERT. Taken together, these results demonstrate that the interaction of the Hsp90-p23 complex with hTERT is critical for regulation of the nuclear localization of telomerase, and that down-regulation of hTERT by curcumin involves dissociating the binding of hTERT with p23. Thus, inhibition of nuclear translocation of hTERT by curcumin may provide new perspectives for regulation of telomerase activity during tumorigenic progression.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , HSP90 Heat-Shock Proteins/drug effects , Intramolecular Oxidoreductases/drug effects , Telomerase/drug effects , Cell Line, Tumor , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Intramolecular Oxidoreductases/metabolism , Prostaglandin-E Synthases , Protein Transport/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism
20.
Proc Natl Acad Sci U S A ; 106(51): 21807-12, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19995978

ABSTRACT

The arachidonic acid (AA) cascade produces eicosanoids, such as prostaglandins (PGs), that regulate physiological and pathological functions. Although various nonsteroidal anti-inflammatory drugs have been developed, blocking upstream components (cyclooxygenase-1 and -2) of the AA cascade leads to severe side effects, including gastrointestinal ulcers and cardiovascular events, respectively, due to the complexity of the AA cascade. Here, using an AA cascade-targeted lipidomics approach, we report that microsomal PGE synthase 1 (mPGES-1) plays a key role in experimental autoimmune encephalomyelitis (EAE). Eicosanoids (mainly PGD(2)) are produced constitutively in the spinal cord of naive mice. However, in EAE lesions, the PGE(2) pathway is favored and the PGD(2), PGI(2), and 5-lipoxygenase pathways are attenuated. Furthermore, mPGES-1(-/-) mice showed less severe symptoms of EAE and lower production of IL-17 and IFN-gamma than mPGES-1(+/+) mice. Expression of PGE(2) receptors (EP1, EP2, and EP4) was elevated in EAE lesions and correlated with clinical symptoms. Immunohistochemistry on central nervous systems of EAE mice and multiple sclerosis (MS) patients revealed overt expression of mPGES-1 protein in microglia/macrophages. Thus, the mPGES-1-PGE(2)-EPs axis of the AA cascade may exacerbate EAE pathology. Our findings have important implications for the design of therapies for MS.


Subject(s)
Dinoprostone/metabolism , Intramolecular Oxidoreductases/drug effects , Lipids , Multiple Sclerosis/drug therapy , Animals , Arachidonic Acid/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Humans , Immunohistochemistry , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/metabolism , Prostaglandin-E Synthases
SELECTION OF CITATIONS
SEARCH DETAIL
...