Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Dig Dis Sci ; 64(9): 2404-2415, 2019 09.
Article in English | MEDLINE | ID: mdl-30830525

ABSTRACT

Zinc is an essential micronutrient for normal organ function, and dysregulation of zinc metabolism has been implicated in a wide range of diseases. Emerging evidence has revealed that zinc transporters play diverse roles in cellular homeostasis and function by regulating zinc trafficking via organelles or the plasma membrane. In the gastrointestinal tract, zinc deficiency leads to diarrhea and dysfunction of intestinal epithelial cells. Studies also showed that zinc transporters are very important in intestinal epithelial homeostasis. In this review, we describe the physiological roles of zinc transporters in intestinal epithelial functions and relevance of zinc transporters in gastrointestinal diseases.


Subject(s)
Cation Transport Proteins/metabolism , Epithelium/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Zinc/metabolism , Animals , Carrier Proteins/metabolism , Cation Transport Proteins/genetics , Gastrointestinal Diseases/genetics , Humans , Intestinal Absorption , Paneth Cells/metabolism , Risk Factors , Stem Cells/metabolism
2.
PLoS Genet ; 12(10): e1006349, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27736879

ABSTRACT

Zinc transporters play a critical role in spatiotemporal regulation of zinc homeostasis. Although disruption of zinc homeostasis has been implicated in disorders such as intestinal inflammation and aberrant epithelial morphology, it is largely unknown which zinc transporters are responsible for the intestinal epithelial homeostasis. Here, we show that Zrt-Irt-like protein (ZIP) transporter ZIP7, which is highly expressed in the intestinal crypt, is essential for intestinal epithelial proliferation. Mice lacking Zip7 in intestinal epithelium triggered endoplasmic reticulum (ER) stress in proliferative progenitor cells, leading to significant cell death of progenitor cells. Zip7 deficiency led to the loss of Olfm4+ intestinal stem cells and the degeneration of post-mitotic Paneth cells, indicating a fundamental requirement for Zip7 in homeostatic intestinal regeneration. Taken together, these findings provide evidence for the importance of ZIP7 in maintenance of intestinal epithelial homeostasis through the regulation of ER function in proliferative progenitor cells and maintenance of intestinal stem cells. Therapeutic targeting of ZIP7 could lead to effective treatment of gastrointestinal disorders.


Subject(s)
Cation Transport Proteins/genetics , Cell Proliferation/genetics , Endoplasmic Reticulum Stress/genetics , Zinc/metabolism , Animals , Apoptosis/genetics , Cation Transport Proteins/biosynthesis , Endoplasmic Reticulum/genetics , Epithelial Cells/metabolism , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/metabolism , Gene Knockout Techniques , Homeostasis , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Mice , Organoids/growth & development , Paneth Cells/metabolism , Stem Cells/metabolism
3.
J Neurochem ; 145(6): 474-488, 2018 06.
Article in English | MEDLINE | ID: mdl-29500815

ABSTRACT

Sepsis-associated encephalopathy (SAE), characterized as diffuse brain dysfunction and neurological manifestations secondary to sepsis, is a common complication in critically ill patients and can give rise to poor outcome, but understanding the molecular basis of this disorder remains a major challenge. Given the emerging role of G protein-coupled receptor 2 (GRK2), first identified as a G protein-coupled receptor (GPCR) regulator, in the regulation of non-G protein-coupled receptor-related molecules contributing to diverse cellular functions and pathology, including inflammation, we tested the hypothesis that GRK2 may be linked to the neuropathogenesis of SAE. When mouse MG6 microglial cells were challenged with lipopolysaccharide (LPS), GRK2 cytosolic expression was highly up-regulated. The ablation of GRK2 by small interfering RNAs (siRNAs) prevented an increase in intracellular reactive oxygen species generation in LPS-stimulated MG6 cells. Furthermore, the LPS-induced up-regulation of inducible nitric-oxide synthase expression and increase in nitric oxide production were negated by GRK2 inhibitor or siRNAs. However, GRK2 inhibition was without effect on overproduction of tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß in LPS-stimulated MG cells. In mice with cecal ligation and puncture-induced sepsis, treatment with GRK2 inhibitor reduced high levels of oxidative and nitrosative stress in the mice brains, where GRK2 expression was up-regulated, alleviated neurohistological damage observed in cerebral cortex sections, and conferred a significant survival advantage to CLP mice. Altogether, these results uncover the novel role for GRK2 in regulating cellular oxidative and nitrosative stress during inflammation and suggest that GRK2 may have a potential as an intriguing therapeutic target to prevent or treat SAE.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Oxidative Stress , Reactive Oxygen Species/metabolism , Sepsis-Associated Encephalopathy/pathology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cytokines/biosynthesis , Enzyme Inhibitors/therapeutic use , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/enzymology , Nitric Oxide Synthase Type II/metabolism , RNA, Small Interfering/pharmacology , Sepsis-Associated Encephalopathy/complications , Sepsis-Associated Encephalopathy/drug therapy , Up-Regulation/drug effects
4.
Crit Care Med ; 46(5): e435-e442, 2018 05.
Article in English | MEDLINE | ID: mdl-29406423

ABSTRACT

OBJECTIVES: Inflammation and apoptosis are decisive mechanisms for the development of end-organ injury in sepsis. Activator protein-1 may play a key role in regulating expression of harmful genes responsible for the pathophysiology of septic end-organ injury along with the major transcription factor nuclear factor-κB. We investigated whether in vivo introduction of circular dumbbell activator protein-1 decoy oligodeoxynucleotides can provide benefits for reducing septic end-organ injury. DESIGN: Laboratory and animal/cell research. SETTINGS: University research laboratory. SUBJECTS: Male BALB/c mice (8-10 wk old). INTERVENTIONS: Activator protein-1 decoy oligodeoxynucleotides were effectively delivered into tissues of septic mice in vivo by preparing into a complex with atelocollagen given 1 hour after surgery. MATERIALS AND MAIN RESULTS: Polymicrobial sepsis was induced by cecal ligation and puncture in mice. Activator protein-1 decoy oligodeoxynucleotide transfection inhibited abnormal production of proinflammatory and chemotactic cytokines after cecal ligation and puncture. Histopathologic changes in lung, liver, and kidney tissues after cecal ligation and puncture were improved by activator protein-1 decoy oligodeoxynucleotide administration. When activator protein-1 decoy oligodeoxynucleotides were given, apoptosis induction was strikingly suppressed in lungs, livers, kidneys, and spleens of cecal ligation and puncture mice. These beneficial effects of activator protein-1 decoy oligodeoxynucleotides led to a significant survival advantage in mice after cecal ligation and puncture. Apoptotic gene profiling indicated that activator protein-1 activation was involved in the up-regulation of many of proapoptotic and antiapoptotic genes in cecal ligation and puncture-induced sepsis. CONCLUSIONS: Our results indicate a detrimental role of activator protein-1 in the sepsis pathophysiology and the potential usefulness of activator protein-1 decoy oligodeoxynucleotides for the prevention and treatment of septic end-organ failure.


Subject(s)
Oligodeoxyribonucleotides/therapeutic use , Sepsis/therapy , Transcription Factor AP-1/therapeutic use , Transfection/methods , Animals , Apoptosis , Cytokines/blood , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Male , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/genetics , Real-Time Polymerase Chain Reaction , Sepsis/genetics , Sepsis/mortality , Sepsis/pathology , Transcription Factor AP-1/genetics
5.
Biochem Biophys Res Commun ; 493(3): 1342-1348, 2017 11 25.
Article in English | MEDLINE | ID: mdl-28970068

ABSTRACT

Aberrant activation of Wnt signaling plays a pivotal role in the development of human cancers including colon cancer. Small compounds that regulate Wnt signaling are attractive candidate for the colon cancer therapy. Here, we showed that SKL2001, which has been identified as an activator for Wnt signaling by disrupting the Axin/ß-Catenin complex, negatively regulates growth of colon cancer spheroids cultured in the 3D condition that simulates tumor microenvironment in vivo. SKL2001 inhibited proliferation of colon cancer cells cultured in 3D spheroid and induced them accumulation in the G0/G1 phase of the cell cycle with a reduced c-myc level. To examine the potential of arrested cells to recover, colon cancer spheroids that were treated with SKL2001 were then cultured in the SKL2001-free medium. We found that SKL2001-treated cells were resumed cell cycle progression and proliferated in the SKL2001-free medium. Notably, SKL2001 facilitated round-shape spheroid formation. This was associated with upregulated expressions of E-cadherin and ß-Catenin. These findings suggest that SKL2001 can suppress colon cancer spheroid growth through regulating cell cycle progression and cadherin/catenin mediated cell-cell contact.


Subject(s)
Cadherins/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Imidazoles/pharmacology , Isoxazoles/pharmacology , beta Catenin/metabolism , Antigens, CD , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Colonic Neoplasms/metabolism , HCT116 Cells , Humans , Spheroids, Cellular/drug effects , Tumor Cells, Cultured , Tumor Microenvironment
6.
Toxicol Appl Pharmacol ; 334: 55-65, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28887131

ABSTRACT

Prostacyclin (PGI2) serves as a protective, anti-inflammatory mediator and PGI2 mimetics may be useful as a hepatoprotective agent. We examined whether two PGI2 mimetics, ONO-1301 and beraprost, are beneficial in acute liver injury and attempted to delineate the possible mechanism underlying the hepatoprotective effect. Acute liver injury was induced by lipopolysaccharide/d-galactosamine (LPS/GalN) in mice. Mice were given an intraperitoneal injection of PGI2 mimetics 1h before LPS/GalN challenge. Both ONO-1301 and beraprost significantly declined the LPS/GalN-induced increase in serum aminotransferase activity. ONO-1301 and, to a lesser extent, beraprost inhibited hepatic gene expression levels of pro-inflammatory cytokines, which were sharply elevated by LPS/GalN. The hepatoprotective effects of ONO-1301, to a lesser extent, of beraprost were also supported by liver histopathological examinations. The PGI2 receptor antagonist CAY10441 abrogated their hepatoprotective effects. The mechanisms behind the benefit of PGI2 mimetics in reducing LPS/GalN-induced liver injury involved, in part, their suppressive effects on increased generation of reactive oxygen species (ROS), since their ability to prevent LPS/GalN-induced hepatic apoptosis was mimicked by the antioxidant N-acetyl-l-cysteine. They significantly diminished LPS/GalN-induced activation of signal transducers and activators of transcription 3 (STAT3) in liver tissues, an effect which was highly associated with their hepatoprotective effects. We indicate that IP receptor activation with PGI2 mimetics can rescue the damage in the liver induced by LPS/GalN by undermining activation of STAT3 and leading to a lower production of ROS. Our findings point to PGI2 mimetics, especially ONO-1301, as a potential novel therapeutic modality for the treatment of acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Epoprostenol/analogs & derivatives , Galactosamine/toxicity , Lipopolysaccharides/toxicity , Pyridines/pharmacology , Animals , Benzyl Compounds/pharmacology , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Epoprostenol/pharmacology , Galactosamine/administration & dosage , Gene Expression Regulation , Imidazoles/pharmacology , Lipopolysaccharides/administration & dosage , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Prostaglandins I/chemistry , Prostaglandins I/pharmacology , Reactive Oxygen Species , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
7.
Am J Pathol ; 185(1): 162-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447055

ABSTRACT

Although phenotypically polarized macrophages are now generally classified into two major subtypes termed proinflammatory M1 and anti-inflammatory M2 macrophages, a contributory role of lung M2 macrophages in the pathophysiological features of acute lung injury is not fully understood. Herein, we show in an endotoxemic murine model that M2 macrophages serve as key anti-inflammatory cells that play a regulatory role in the severity of lung injury. To study whether M2 macrophages can modify inflammation, we depleted M2 macrophages from lungs of CD206-diphtheria toxin (DT) receptor transgenic (Tg) mice during challenge with lipopolysaccharide. The i.p. administration of DT depleted CD206-positive cells in bronchoalveolar lavage fluid. The use of M2 macrophage markers Ym1 and arginase-1 identified pulmonary CD206-positive cells as M2 macrophages. A striking increase in neutrophils in bronchoalveolar lavage fluid cell contents was found in DT-treated CD206-DT receptor Tg mice. In CD206-DT receptor Tg mice given DT, endotoxin challenge exaggerated lung inflammation, including up-regulation of proinflammatory cytokines and increased histological lung damage, but the endotoxemia-induced increase in NF-κB activity was significantly reduced, suggesting that M2 phenotype-dependent counteraction of inflammatory insult cannot be attributed to the inhibition of the NF-κB pathway. Our results indicate a critical role of CD206-positive pulmonary macrophages in triggering inflammatory cascade during endotoxemic lung inflammation.


Subject(s)
Acute Lung Injury/metabolism , Endotoxemia/pathology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/metabolism , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cell Membrane/metabolism , Chromosomes, Artificial, Bacterial , Endotoxemia/metabolism , Exons , Humans , Inflammation/pathology , Lipopolysaccharides , Lung/metabolism , Macrophages/cytology , Mannose Receptor , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Neutrophils/cytology , Phenotype , RNA, Messenger/metabolism , Rabbits , Transcription Factor AP-1/metabolism
8.
J Pharmacol Exp Ther ; 354(3): 240-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26136420

ABSTRACT

Macrophages are a well recognized player of both innate and adaptive immunity and have emerged as a key regulator of systemicmetabolism, hematopoiesis, vasculogenesis, apoptosis, malignancy, and reproduction. Such pleiotropic roles of macrophages are mirrored by their protean features. Upon environmental. challenges, macrophages redistribute and differentiate in situ and contribute to the multiple disease states by exerting protective and pathogenic effects. The environmental challenges include cytokines, chemokines, lipid mediators, and extrinsic insults, such as food and pathogenic bacteria. In addition, homeostasis and the activation state of macrophages are influenced by various metabolites from a commensal microbe that colonizes epithelial and mucosal surfaces, such as the lungs, intestines, and skin. In this review, we describe macrophage differentiation, polarization, and various functions in chronic disease states, including chronic inflammatory bowel disease, tumorigenesis, metabolism and obesity, and central nervous system demyelinating disorders. Controlling the macrophage dynamics to affect the pathologic states is considered to be an important therapeutic approach for many clinical disorders involving chronic inflammation.


Subject(s)
Chronic Disease/therapy , Inflammation/pathology , Macrophages/pathology , Animals , Cell Differentiation/immunology , Cell Differentiation/physiology , Humans , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation/immunology , Macrophage Activation/physiology , Macrophages/immunology , Macrophages/metabolism
9.
Crit Care Med ; 43(11): e508-20, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26468714

ABSTRACT

OBJECTIVES: The calcium sensitizer levosimendan is used in treatment of decompensated heart failure and may also exhibit anti-inflammatory properties. We examined whether treatment with levosimendan is substantially beneficial in mice with cecal ligation and puncture-induced polymicrobial sepsis, and its arbitration mechanism was explored in the mouse macrophage cell line RAW264.7. DESIGN: Laboratory and animal/cell research. SETTING: University research laboratory. SUBJECTS: BALB/c mice (8-10 wk old) and mouse macrophage cell line RAW264.7 cells. INTERVENTIONS: Levosimendan (0.5 µg/kg/min) was administered to mice through an osmotic pump that was implanted into the peritoneal cavity immediately following surgery. In RAW264.7 cells, levosimendan was added to the culture medium 30 minutes before lipopolysaccharide. MEASUREMENTS AND MAIN RESULTS: When levosimendan was continuously administered to cecal ligation and puncture-induced septic mice, a significant improvement of left ventricular function was found without any change in heart rate, and hypotension was significantly mitigated. Furthermore, levosimendan conferred substantial protection against sepsis-associated inflammation in mice, as indicated by reduced lung injury and decreased blood proinflammatory and chemotactic cytokine levels. These beneficial effects of levosimendan led to a significant improvement of survival in mice after cecal ligation and puncture. In endotoxin-stimulated RAW264.7 macrophages, treatment with levosimendan and pimobendan suppressed overproduction of proinflammatory and chemotactic cytokines. Levosimendan and pimobendan were without effect on activation of the nuclear factor-κB, mitogen-activated protein kinase, and Akt pathways. Instead, levosimendan and pimobendan prevented high mobility group box 1 release from the nucleus to the extracellular space in macrophages. This was associated with inhibition of the Rho kinase signaling pathway. The elevated serum high mobility group box 1 levels in cecal ligation and puncture-induced septic mice were also inhibited by continued administration of levosimendan and pimobendan. CONCLUSIONS: We define a novel mechanism for the anti-inflammatory action of levosimendan and suggest that the pharmacological profiles of levosimendan as both an inotrope and an anti-inflammatory agent could contribute to its clinical benefit in patients with sepsis with heart problems.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Hydrazones/pharmacology , Macrophages/drug effects , Pyridazines/pharmacology , Sepsis/drug therapy , Sepsis/pathology , Animals , Biopsy, Needle , Blotting, Western , Cecum/surgery , Cells, Cultured , Cytokines/drug effects , Disease Models, Animal , Echocardiography/methods , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Injections, Intravenous , Ligation/methods , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Random Allocation , Real-Time Polymerase Chain Reaction , Reference Values , Sepsis/mortality , Simendan , Statistics, Nonparametric , Survival Rate
10.
J Surg Res ; 193(2): 874-87, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25291964

ABSTRACT

BACKGROUND: Cardiac dysfunction is a frequent and severe complication of septic shock and contributes to the high mortality of sepsis. Although several mechanisms have been suspected to be responsible for sepsis-associated cardiac dysfunction, the precise cause(s) remains unclear to date. MATERIALS AND METHODS: We tested the hypothesis that cardiac fibroblasts may play a critical role as a disease modifier involved in sepsis-associated cardiac dysfunction. Human cardiac fibroblasts (HCFs) cultured in vitro were exposed to lipopolysaccharide (LPS). Changes in cardiac morphology and function were assessed in mice with cecal ligation and puncture-induced sepsis. RESULTS: In LPS-stimulated HCFs, messenger RNA and protein levels of proinflammatory molecules, including tumor necrosis factor-α, interleukin-1ß, interleukin-6, and monocyte chemoattractant protein-1, were strikingly upregulated. LPS also increased expression and activity of matrix metalloproteinase (MMP)-9, but not MMP-2. LPS-induced expression of α-smooth muscle actin, a classical marker for myoblast differentiation, which was abrogated when MMP-9 small interfering RNA was transfected into HCFs. High gene expression levels of proinflammatory cytokines and MMP-9 were observed in the heart tissues of cecal ligation and puncture-induced septic mice. Histology sections of the hearts from septic mice showed perivascular and interstitial cardiac fibrosis, and echocardiography demonstrated that septic mice had profound cardiac dysfunction. The broad-spectrum MMP inhibitor ONO-4817 significantly alleviated these histologic and functional changes during the acute phase. CONCLUSIONS: We suggest that cardiac fibroblasts are of pathogenetic importance in inflammation and fibrosis in the heart during sepsis, leading to cardiac dysfunction that would affect the outcome of sepsis syndrome.


Subject(s)
Fibroblasts/physiology , Heart/physiopathology , Shock, Septic/physiopathology , Actins/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Fibrosis , History, Ancient , Humans , Lipopolysaccharides , Male , Matrix Metalloproteinases/metabolism , Mice, Inbred BALB C , Myocardium/pathology , NF-kappa B/metabolism , Phenyl Ethers , Shock, Septic/pathology
11.
J Pharmacol Exp Ther ; 349(2): 199-208, 2014 May.
Article in English | MEDLINE | ID: mdl-24570070

ABSTRACT

G protein-coupled receptor kinase 2 (GRK2) participates together with ß-arrestins in the regulation of G protein-coupled receptor signaling, but emerging evidence suggests that GRK2 can interact with a growing number of proteins involved in signaling mediated by other membrane receptor families under various pathologic conditions. We tested the hypothesis that GRK2 may be an important contributor to vascular endothelial dysfunction in diabetes. Human umbilical venous endothelial cells (HUVECs) were exposed to high glucose and high insulin (HG/HI) to mimic insulin-resistant diabetic conditions. GRK2 expression and membrane translocation were up-regulated under HG/HI conditions. HG/HI did not modify activation of Akt or endothelial nitric-oxide synthase (eNOS), but GRK2 inhibitor or small interfering RNA (siRNA) resulted in an increase in Akt and eNOS activation in HUVECs exposed to HG/HI. Extracellular signal-regulated kinase 1/2 (ERK1/2) activation was increased after exposure to HG/HI, which was prevented by GRK2 inhibitor or siRNA. ERK1/2-mediated GRK2 phosphorylation at Ser-670 confirmed that ERK1/2 participated in a negative feedback regulatory loop. In human embryonic kidney 293T cells that overexpressed GRK2, Akt activity was unchanged, whereas ERK1/2 activity was raised. The effect of GRK inhibitor treatment on Akt/eNOS signaling was associated with membrane translocation of ß-arrestin 2. The experiments with ß-arrestin 2 siRNA showed that ß-arrestin 2 may act as a positive modulator of Akt/eNOS signaling. Our studies reveal that GRK2, which is up-regulated by HG/HI, leads to a tonic inhibition of the insulin Akt/eNOS pathway in endothelial cells. We provide new insights into the pathogenesis of diabetes-associated vascular endothelial dysfunction.


Subject(s)
Endothelium, Vascular/metabolism , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Glucose/metabolism , Insulin/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Arrestins/metabolism , Diabetes Mellitus/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Insulin Resistance , Nitrofurans/pharmacology , Protein Transport , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Vinyl Compounds/pharmacology , beta-Arrestin 2 , beta-Arrestins
12.
Cell Rep ; 43(7): 114490, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990720

ABSTRACT

Although oral tolerance is a critical system in regulating allergic disorders, the mechanisms by which dietary factors regulate the induction and maintenance of oral tolerance remain unclear. To address this, we explored the differentiation and function of various immune cells in the intestinal immune system under fasting and ad libitum-fed conditions before oral ovalbumin (OVA) administration. Fasting mitigated OVA-specific Treg expansion, which is essential for oral tolerance induction. This abnormality mainly resulted from functional defects in the CX3CR1+ cells responsible for the uptake of luminal OVA and reduction of tolerogenic CD103+ dendritic cells. Eventually, fasting impaired the preventive effect of oral OVA administration on asthma and allergic rhinitis development. Specific food ingredients, namely carbohydrates and arginine, were indispensable for oral tolerance induction by activating glycolysis and mTOR signaling. Overall, prior food intake and nutritional signals are critical for maintaining immune homeostasis by inducing tolerance to ingested food antigens.


Subject(s)
Arginine , Dendritic Cells , Immune Tolerance , Ovalbumin , T-Lymphocytes, Regulatory , TOR Serine-Threonine Kinases , Animals , Arginine/metabolism , T-Lymphocytes, Regulatory/immunology , Ovalbumin/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Administration, Oral , CX3C Chemokine Receptor 1/metabolism , Intestines/immunology , Antigens, CD/metabolism , Integrin alpha Chains/metabolism , Sugars/metabolism , Glycolysis , Fasting , Signal Transduction , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Female
13.
J Biol Chem ; 286(46): 40255-65, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21917916

ABSTRACT

The human SLC39A13 gene encodes ZIP13, a member of the LZT (LIV-1 subfamily of ZIP zinc transporters) family. The ZIP13 protein is important for connective tissue development, and its loss of function is causative for the spondylocheiro dysplastic form of Ehlers-Danlos syndrome. However, this protein has not been characterized in detail. Here we report the first detailed biochemical characterization of the human ZIP13 protein using its ectopic expressed and the purified recombinant protein. Protease accessibility, microscopic, and computational analyses demonstrated that ZIP13 contains eight putative transmembrane domains and a unique hydrophilic region and that it resides with both its N and C termini facing the luminal side on the Golgi. Analyses including cross-linking, immunoprecipitation, Blue Native-PAGE, and size-exclusion chromatography experiments indicated that the ZIP13 protein may form a homo-dimer. We also demonstrated that ZIP13 mediates zinc influx, as assessed by monitoring the expression of the metallothionein gene and by detecting the intracellular zinc level with a zinc indicator, FluoZin-3. Our data indicate that ZIP13 is a homo-dimerized zinc transporter that possesses some domains that are not found in other LZT family members. This is the first biochemical characterization of the physiologically important protein ZIP13 and the demonstration of homo-dimerization for a mammalian ZIP zinc transporter family member. This biochemical characterization of the human ZIP13 protein provides important information for further investigations of its structural characteristics and function.


Subject(s)
Cation Transport Proteins/metabolism , Ehlers-Danlos Syndrome/metabolism , Protein Multimerization , Zinc/metabolism , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Line , Ehlers-Danlos Syndrome/genetics , Humans , Ion Transport/genetics , Polycyclic Compounds/chemistry , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Zinc/chemistry
14.
Biosci Biotechnol Biochem ; 76(3): 447-53, 2012.
Article in English | MEDLINE | ID: mdl-22451383

ABSTRACT

Lachrymatory factor synthase (LFS), an enzyme essential for the synthesis of the onion lachrymatory factor (propanethial S-oxide), was identified in 2002. This was the first reported enzyme involved in the production of thioaldehyde S-oxides via an intra-molecular H(+) substitution reaction, and we therefore attempted to identify the catalytic amino acid residues of LFS as the first step in elucidating the unique catalytic reaction mechanism of this enzyme. A comparison of the LFS cDNA sequences among lachrymatory Allium plants, a deletion analysis and site-directed mutagenesis enabled us to identify two amino acids (Arg71 and Glu88) that were indispensable to the LFS activity. Homology modeling was performed for LFS/23-169 on the basis of the template structure of a pyrabactin resistance 1-like protein (PYL) which had been selected from a BLASTP search on SWISS-MODEL against LFS/23-169. We identified in the modeled structure of LFS a pocket corresponding to the ligand-binding site in PYL, and Arg71 and Glu88 were located in this pocket.


Subject(s)
Allium/enzymology , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Allium/genetics , Amino Acid Sequence , Biocatalysis , Cloning, Molecular , DNA, Complementary/genetics , Intramolecular Oxidoreductases/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Sequence Homology, Amino Acid
15.
Int Immunol ; 22(5): 375-86, 2010 May.
Article in English | MEDLINE | ID: mdl-20215335

ABSTRACT

Zinc (Zn) is an essential trace metal required by many enzymes and transcription factors for their activity or the maintenance of their structure. Zn has a variety of effects in the immune responses and inflammation, although it has not been well known how Zn affects these reactions on the molecular basis. We here showed that Zn suppresses T(h)17-mediated autoimmune diseases at lest in part by inhibiting the development of T(h)17 cells via attenuating STAT3 activation. In mice injected with type II collagen to induce arthritis, Zn treatment inhibited T(h)17 cell development. IL-6-mediated activation of STAT3 and in vitro T(h)17 cell development were all suppressed by Zn. Importantly, Zn binding changed the alpha-helical secondary structure of STAT3, disrupting the association of STAT3 with JAK2 kinase and with a phospho-peptide that included a STAT3-binding motif from the IL-6 signal transducer gp130. Thus, we conclude that Zn suppresses STAT3 activation, which is a critical step for T(h)17 development.


Subject(s)
Arthritis, Experimental/drug therapy , Interleukin-17/immunology , STAT3 Transcription Factor/antagonists & inhibitors , Th17 Cells/drug effects , Th17 Cells/immunology , Zinc/pharmacology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Th17 Cells/cytology , Th17 Cells/metabolism
16.
Sci Rep ; 11(1): 24154, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921186

ABSTRACT

Sepsis is a systemic reaction to an infection and resulting in excessive production of inflammatory cytokines and chemokines. It sometimes results in septic shock. The present study aimed to identify quinolone antibiotics that can reduce tumor necrosis factor alpha (TNFα) production and to elucidate mechanisms underlying inhibition of TNFα production. We identified quinolone antibiotics reduced TNFα production in lipopolysaccharide (LPS)-stimulated THP-1 cells. Sitafloxacin (STFX) is a broad-spectrum antibiotic of the quinolone class. STFX effectively suppressed TNFα production in LPS-stimulated THP-1 cells in a dose-dependent manner and increased extracellular signal-regulated kinase (ERK) phosphorylation. The percentage of intracellular TNFα increased in LPS-stimulated cells with STFX compared with that in LPS-stimulated cells. TNFα converting enzyme (TACE) released TNFα from the cells, and STFX suppressed TACE phosphorylation and activity. To conclude, one of the mechanisms underlying inhibition of TNFα production in LPS-stimulated THP-1 cells treated with STFX is the inhibition of TNFα release from cells via the suppression of TACE phosphorylation and activity. STFX may kill bacteria and suppress inflammation. Therefore, it can be effective for sepsis treatment.


Subject(s)
ADAM17 Protein/metabolism , Fluoroquinolones/pharmacology , Lipopolysaccharides/toxicity , Tumor Necrosis Factor-alpha/metabolism , Humans , Phosphorylation/drug effects , THP-1 Cells
17.
Nat Commun ; 12(1): 2105, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833232

ABSTRACT

Intestinal microbiota-derived metabolites have biological importance for the host. Polyamines, such as putrescine and spermidine, are produced by the intestinal microbiota and regulate multiple biological processes. Increased colonic luminal polyamines promote longevity in mice. However, no direct evidence has shown that microbial polyamines are incorporated into host cells to regulate cellular responses. Here, we show that microbial polyamines reinforce colonic epithelial proliferation and regulate macrophage differentiation. Colonisation by wild-type, but not polyamine biosynthesis-deficient, Escherichia coli in germ-free mice raises intracellular polyamine levels in colonocytes, accelerating epithelial renewal. Commensal bacterium-derived putrescine increases the abundance of anti-inflammatory macrophages in the colon. The bacterial polyamines ameliorate symptoms of dextran sulfate sodium-induced colitis in mice. These effects mainly result from enhanced hypusination of eukaryotic initiation translation factor. We conclude that bacterial putrescine functions as a substrate for symbiotic metabolism and is further absorbed and metabolised by the host, thus helping maintain mucosal homoeostasis in the intestine.


Subject(s)
Colon/metabolism , Escherichia coli/metabolism , Intestinal Mucosa/metabolism , Peptide Initiation Factors/metabolism , Putrescine/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Epithelial Cells/metabolism , Female , Gastrointestinal Microbiome/physiology , Homeostasis , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Eukaryotic Translation Initiation Factor 5A
18.
J Immunol Res ; 2019: 8396878, 2019.
Article in English | MEDLINE | ID: mdl-30984791

ABSTRACT

Intestinal epithelial cells cover the surface of the intestinal tract. The cells are important for preserving the integrity of the mucosal barriers to protect the host from luminal antigens and pathogens. The mucosal barriers are maintained by the continuous and rapid self-renewal of intestinal epithelial cells. Defects in the self-renewal of these cells are associated with gastrointestinal diseases, including inflammatory bowel diseases and diarrhea. Zinc is an essential trace element for living organisms, and zinc deficiency is closely linked to the impaired mucosal integrity. Recent evidence has shown that zinc transporters contribute to the barrier function of intestinal epithelial cells. In this review, we describe the recent advances in understanding the role of zinc and zinc transporters in the barrier function and homeostasis of intestinal epithelial cells.


Subject(s)
Carrier Proteins/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Zinc/metabolism , Animals , Biological Transport , Epithelial Cells/immunology , Homeostasis , Humans , Intestinal Mucosa/immunology , Intestines/immunology , Intestines/pathology , Mice
19.
Front Pharmacol ; 10: 59, 2019.
Article in English | MEDLINE | ID: mdl-30778300

ABSTRACT

G protein-coupled receptor kinase 2 (GRK2) is a ubiquitous member of the GRK family that restrains cellular activation by G protein-coupled receptor (GPCR) phosphorylation leading to receptor desensitization and internalization, but has been identified to regulate a variety of signaling molecules, among which may be associated with inflammation. In this study, we attempted to establish the regulatory role of GRK2 in the Toll-like receptor (TLR) signaling pathway for inducible nitric oxide synthase (iNOS) expression in microglial cells. When mouse MG6 cells were stimulated with the TLR4 ligands lipopolysaccharide (LPS) and paclitaxel, we found that interferon regulatory factor 1 (IRF1) protein expression and activation was upregulated, transcription of interferon-ß (IFN-ß) was accelerated, induction/activation of STAT1 and activation of STAT3 were promoted, and subsequently iNOS expression was upregulated. The ablation of GRK2 by small interfering RNAs (siRNAs) not only eliminated TLR4-mediated upregulation of IRF1 protein expression and nuclear translocation but also suppressed the activation of the STAT pathway, resulting in negating the iNOS upregulation. The TLR3-mediated changes in IRF1 and STAT1/3, leading to iNOS induction, were also abrogated by siRNA knockdown of GRK2. Furthermore, transfection of GRK2 siRNA blocked the exogenous IFN-ß supplementation-induced increases in phosphorylation of STAT1 as well as STAT3 and abrogated the augmentation of iNOS expression in the presence of exogenous IFN-ß. Taken together, our results show that GRK2 regulates the activation of IRF1 as well as the activation of the STAT pathway, leading to upregulated transcription of iNOS in activated microglial cells. Modulation of the TLR signaling pathway via GRK2 in microglia may be a novel therapeutic target for treatment of neuroinflammatory disorders.

20.
Sci Rep ; 9(1): 10842, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346193

ABSTRACT

Zinc (Zn) is an essential nutrient and its deficiency causes immunodeficiency and skin disorders. Various cells including mast cells release Zn-containing granules when activated; however, the biological role of the released Zn is currently unclear. Here we report our findings that Zn transporter ZnT2 is required for the release of Zn from mast cells. In addition, we found that Zn and mast cells induce IL-6 production from inflammatory cells such as skin fibroblasts and promote wound healing, a process that involves inflammation. Zn induces the production of a variety of pro-inflammatory cytokines including IL-6 through signaling pathways mediated by the Zn receptor GPR39. Consistent with these findings, wound healing was impaired in mice lacking IL-6 or GPR39. Thus, our results show that Zn and mast cells play a critical role in wound healing through activation of the GPR39/IL-6 signaling axis.


Subject(s)
Cation Transport Proteins/metabolism , Interleukin-6/metabolism , Mast Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Wound Healing/physiology , Animals , Cell Line , Cells, Cultured , Mast Cells/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL