Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
J Cell Physiol ; 237(2): 1455-1470, 2022 02.
Article in English | MEDLINE | ID: mdl-34705285

ABSTRACT

Spinal cord injury (SCI) causes cell death and consequently the breakdown of axons and myelin. The accumulation of myelin debris at the lesion site induces inflammation and blocks axonal regeneration. Hematogenous macrophages contribute to the removal of myelin debris. In this study, we asked how the inflammatory state of macrophages affects their ability to phagocytose myelin. Bone marrow-derived macrophages (BMDM) and Raw264.7 cells were stimulated with lipopolysaccharides (LPS) or interferon gamma (IFNγ), which induce inflammatory stress, and the endocytosis of myelin was examined. We found that activation of the TLR4-NFκB pathway reduced myelin uptake by BMDM, while IFNγ-Jak/STAT1 signaling did not. Since bile acids regulate lipid metabolism and in some cases reduce inflammation, our second objective was to investigate whether myelin clearance could be improved with taurolithocholic acid (TLCA), tauroursodeoxycholic acid or hyodeoxycholic acid. In BMDM only TLCA rescued myelin phagocytosis, when this activity was suppressed by LPS. Inhibition of protein kinase A blocked the effect of TLCA, while an agonist of the farnesoid X receptor did not rescue phagocytosis, implicating TGR5-PKA signaling in the effect of TLCA. To shed light on the mechanism, we measured whether TLCA affected the expression of CD36, triggering receptor on myeloid cells-2 (TREM2), and Gas6, which are known to be involved in phagocytosis and affected by inflammatory stimuli. Concomitant with an increase in expression of tumour necrosis factor alpha, LPS reduced expression of TREM2 and Gas6 in BMDM, and TLCA significantly diminished this downregulation. These findings suggest that activation of bile acid receptors may be used to improve myelin clearance in neuropathologies.


Subject(s)
Lipopolysaccharides , Taurolithocholic Acid , Humans , Inflammation/pathology , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Myelin Sheath , Phagocytosis , Taurochenodeoxycholic Acid , Taurolithocholic Acid/metabolism , Taurolithocholic Acid/pharmacology
2.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L264-L275, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31800261

ABSTRACT

Duodenogastroesophageal reflux (DGER) is associated with chronic lung disease. Bile acids (BAs) are established markers of DGER aspiration and are important risk factors for reduced post-transplant lung allograft survival by disrupting the organ-specific innate immunity, facilitating airway infection and allograft failure. However, it is unknown whether BAs also affect airway reactivity. We investigated the acute effects of 13 BAs detected in post-lung-transplant surveillance bronchial washings (BW) on airway contraction. We exposed precision-cut slices from human and mouse lungs to BAs and monitored dynamic changes in the cross-sectional luminal area of peripheral airways using video phase-contrast microscopy. We also used guinea pig tracheal rings in organ baths to study BA effects in proximal airway contraction induced by electrical field stimulation. We found that most secondary BAs at low micromolar concentrations strongly and reversibly relaxed smooth muscle and inhibited peripheral airway constriction induced by acetylcholine but not by noncholinergic bronchoconstrictors. Similarly, secondary BAs strongly inhibited cholinergic constrictions in tracheal rings. In contrast, TC-G 1005, a specific agonist of the BA receptor Takeda G protein-coupled receptor 5 (TGR5), did not cause airway relaxation, and Tgr5 deletion in knockout mice did not affect BA-induced relaxation, suggesting that this receptor is not involved. BAs inhibited acetylcholine-induced inositol phosphate synthesis in human airway smooth muscle cells overexpressing the muscarinic M3 receptor. Our results demonstrate that select BAs found in BW of patients with lung transplantation can affect airway reactivity by inhibiting the cholinergic contractile responses of the proximal and peripheral airways, possibly by acting as antagonists of M3 muscarinic receptors.


Subject(s)
Acetylcholine/metabolism , Bile Acids and Salts/pharmacology , Bronchoconstriction/drug effects , Lung/physiopathology , Animals , Bronchoconstrictor Agents/pharmacology , Chenodeoxycholic Acid/pharmacology , Electric Stimulation , Guinea Pigs , Humans , Inositol Phosphates/biosynthesis , Lung/drug effects , Male , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Muscarinic/metabolism , Serotonin/pharmacology , Taurolithocholic Acid/pharmacology , Trachea/drug effects
3.
Arch Toxicol ; 93(8): 2279-2294, 2019 08.
Article in English | MEDLINE | ID: mdl-31300867

ABSTRACT

Taurolithocholate (TLC) is a cholestatic bile salt that induces disinsertion of the canalicular transporter Abcc2 (Mrp2, multidrug resistance-associated protein 2). This internalization is mediated by different intracellular signaling proteins such as PI3K, PKCε and MARCK but the initial receptor of TLC remains unknown. A few G protein-coupled receptors interact with bile salts in hepatocytes. Among them, sphingosine-1 phosphate receptor 2 (S1PR2) represents a potential initial receptor for TLC. The aim of this study was to evaluate the role of this receptor and its downstream effectors in the impairment of Abcc2 function induced by TLC. In vitro, S1PR2 inhibition by JTE-013 or its knockdown by small interfering RNA partially prevented the decrease in Abcc2 activity induced by TLC. Moreover, adenylyl cyclase (AC)/PKA and PI3K/Akt inhibition partially prevented TLC effect on canalicular transporter function. TLC produced PKA and Akt activation, which were blocked by JTE-013 and AC inhibitors, connecting S1PR2/AC/PKA and PI3K/Akt in a same pathway. In isolated perfused rat liver, injection of TLC triggered endocytosis of Abcc2 that was accompanied by a sustained decrease in the bile flow and the biliary excretion of the Abcc2 substrate dinitrophenyl-glutathione until the end of the perfusion period. S1PR2 or AC inhibition did not prevent the initial decay, but they accelerated the recovery of these parameters and the reinsertion of Abcc2 into the canalicular membrane. In conclusion, S1PR2 and the subsequent activation of AC, PKA, PI3K and Akt is partially responsible for the cholestatic effects of TLC through sustained internalization of Abcc2.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenylyl Cyclases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Taurolithocholic Acid/pharmacology , Animals , Cells, Cultured , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver/drug effects , Liver/metabolism , Metabolic Networks and Pathways/drug effects , Organ Culture Techniques , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Rats, Wistar , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Sphingosine-1-Phosphate Receptors/genetics , Taurolithocholic Acid/metabolism
4.
J Biol Chem ; 292(51): 21047-21059, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29042438

ABSTRACT

Acute pancreatitis is a disease associated with inflammation and tissue damage. One protein that protects against acute injury, including ischemic injury to both the kidney and heart, is renalase, which is secreted into the blood by the kidney and other tissues. However, whether renalase reduces acute injury associated with pancreatitis is unknown. Here, we used both in vitro and in vivo murine models of acute pancreatitis to study renalase's effects on this condition. In isolated pancreatic lobules, pretreatment with recombinant human renalase (rRNLS) blocked zymogen activation caused by cerulein, carbachol, and a bile acid. Renalase also blocked cerulein-induced cell injury and histological changes. In the in vivo cerulein model of pancreatitis, genetic deletion of renalase resulted in more severe disease, and administering rRNLS to cerulein-exposed WT mice after pancreatitis onset was protective. Because pathological increases in acinar cell cytosolic calcium levels are central to the initiation of acute pancreatitis, we also investigated whether rRNLS could function through its binding protein, plasma membrane calcium ATPase 4b (PMCA4b), which excretes calcium from cells. We found that PMCA4b is expressed in both murine and human acinar cells and that a PMCA4b-selective inhibitor worsens pancreatitis-induced injury and blocks the protective effects of rRNLS. These findings suggest that renalase is a protective plasma protein that reduces acinar cell injury through a plasma membrane calcium ATPase. Because exogenous rRNLS reduces the severity of acute pancreatitis, it has potential as a therapeutic agent.


Subject(s)
Monoamine Oxidase/metabolism , Pancreas/metabolism , Pancreatitis/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Cell Line , Ceruletide/toxicity , Enzyme Activation/drug effects , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hypertension/etiology , Hypertension/prevention & control , Ligands , Membrane Transport Modulators/pharmacology , Mice , Mice, Knockout , Monoamine Oxidase/blood , Monoamine Oxidase/genetics , Monoamine Oxidase/therapeutic use , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Pancreatitis/pathology , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Plasma Membrane Calcium-Transporting ATPases/chemistry , Plasma Membrane Calcium-Transporting ATPases/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
5.
Biochem Biophys Res Commun ; 500(4): 952-957, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29709472

ABSTRACT

Impairment of mitochondrial biogenesis has been associated with vascular pathophysiology. The G-protein-coupled receptor (TGR5) is an important mediator of bile acid signaling and glucose metabolism. However, the effects of TGR5 on mitochondrial biogenesis in endothelial cells remain elusive. In this study, we found that activation of TGR5 using its specific agonist taurolithocholic acid (TLCA) promoted the expression of PGC-1α, a master regulator of mitochondrial biogenesis in human aortic endothelial cells (HAECs). Additionally, activation of TGR5 increased the expression of PGC-1α target genes, such as NRF1 and TFAM. Indeed, we found that TLCA treatment promoted mitochondrial biogenesis by increasing mitochondrial mass, mitochondrial-to-nuclear DNA (mtDNA/nDNA), COX-Ⅰ expression, and cytochrome c oxidase activity in HAECs. Notably, our results displayed that activation of TGR5 resulted in a functional gain in mitochondria by increasing the rate of respiration and ATP production. Mechanistically, we found that TLCA treatment activated the transcriptional factor CREB by inducing the phosphorylation of CREB at Ser133. Using the PKA/CREB inhibitor H89 abolished the effects of TLCA on PGC-1α, NRF1 and TFAM expression as well as the increase in mtDNA/nDNA and ATP production. These findings suggest that activation of TGR5 promoted mitochondrial biogenesis in endothelial cells, which is mediated by the CREB/PGC-1α signaling pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Endothelial Cells/drug effects , Mitochondria/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, G-Protein-Coupled/genetics , Taurolithocholic Acid/pharmacology , Adenosine Triphosphate/biosynthesis , Cell Line , Cell Respiration , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Isoquinolines/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sulfonamides/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Am J Physiol Gastrointest Liver Physiol ; 311(5): G974-G980, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27686613

ABSTRACT

In the current study, we have characterized the global miRNA expression profile in mouse pancreatic acinar cells and during acute pancreatitis using next-generation RNA sequencing. We identified 324 known and six novel miRNAs that are expressed in mouse pancreatic acinar cells. In the basal state, miR-148a-3p, miR-375-3p, miR-217-5p, and miR-200a-3p were among the most abundantly expressed, whereas miR-24-5p and miR-421-3p were the least abundant. Treatment of acinar cells with caerulein (100 nM) and taurolithocholic acid 3-sulfate [TLC-S (250 µM)] induced numerous changes in miRNA expression profile. In particular, we found significant overexpression of miR-21-3p in acini treated with caerulein and TLC-S. We further looked at the expression of miR-21-3p in caerulein, l-arginine, and caerulein + LPS-induced acute pancreatitis mouse models and found 12-, 21-, and 50-fold increased expression in the pancreas, respectively. In summary, this is the first comprehensive analysis of global miRNA expression profile of mouse pancreatic acinar cells in normal and disease conditions. Our analysis shows that miR-21-3p expression level correlates with the severity of the disease.


Subject(s)
Acinar Cells/metabolism , MicroRNAs/metabolism , Pancreatitis/metabolism , Acinar Cells/drug effects , Animals , Ceruletide/pharmacology , Gene Expression/drug effects , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Mice , MicroRNAs/genetics , Pancreatitis/genetics , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
7.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G999-G1005, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27012769

ABSTRACT

In hepatocytes, cAMP both activates p38 mitogen-activated protein kinase (MAPK) and increases the amount of multidrug resistance-associated protein-2 (MRP2) in the plasma membrane (PM-MRP2). Paradoxically, taurolithocholate (TLC) activates p38 MAPK but decreases PM-MRP2 in hepatocytes. These opposing effects of cAMP and TLC could be mediated via different p38 MAPK isoforms (α and ß) that are activated differentially by upstream kinases (MKK3, MKK4, and MKK6). Thus we tested the hypothesis that p38α MAPK and p38ß MAPK mediate increases and decreases in PM-MRP2 by cAMP and TLC, respectively. Studies were conducted in hepatocytes isolated from C57BL/6 wild-type (WT) and MKK3-knockout (MKK3(-/-)) mice and in a hepatoma cell line (HuH7) that overexpresses sodium-taurocholate cotransporting polypeptide (NTCP) (HuH-NTCP). Cyclic AMP activated MKK3, p38 MAPK, and p38α MAPK and increased PM-MRP2 in WT hepatocytes, but failed to activate p38α MAPK or increase PM-MRP2 in MKK3(-/-) hepatocytes. In contrast to cAMP, TLC activated total p38 MAPK but decreased PM-MRP2, and did not activate MKK3 or p38α MAPK in WT hepatocytes. In MKK3(-/-) hepatocytes, TLC still decreased PM-MRP2 and activated p38 MAPK, indicating that these effects are not MKK3-dependent. Additionally, TLC activated MKK6 in MKK3(-/-) hepatocytes, and small interfering RNA knockdown of p38ß MAPK abrogated TLC-mediated decreases in PM-MRP2 in HuH-NTCP cells. Taken together, these results suggest that p38α MAPK facilitates plasma membrane insertion of MRP2 by cAMP, whereas p38ß MAPK mediates retrieval of PM-MRP2 by TLC.


Subject(s)
Cell Membrane/metabolism , Multidrug Resistance-Associated Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Cyclic AMP/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , Mice , Mice, Inbred C57BL , Multidrug Resistance-Associated Protein 2 , Protein Transport , Taurolithocholic Acid/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics
8.
J Exp Biol ; 218(Pt 20): 3257-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26347551

ABSTRACT

The consensus view of olfactory processing is that the axons of receptor-specific primary olfactory sensory neurons (OSNs) converge to a small subset of glomeruli, thus preserving the odour identity before the olfactory information is processed in higher brain centres. In the present study, we show that two different subsets of ciliated OSNs with different odorant specificities converge to the same glomeruli. In order to stain different ciliated OSNs in the crucian carp Carassius carassius we used two different chemical odorants, a bile salt and a purported alarm substance, together with fluorescent dextrans. The dye is transported within the axons and stains glomeruli in the olfactory bulb. Interestingly, the axons from the ciliated OSNs co-converge to the same glomeruli. Despite intermingled innervation of glomeruli, axons and terminal fields from the two different subsets of ciliated OSNs remained mono-coloured. By 4-6 days after staining, the dye was transported trans-synaptically to separately stained axons of relay neurons. These findings demonstrate that specificity of the primary neurons is retained in the olfactory pathways despite mixed innervation of the olfactory glomeruli. The results are discussed in relation to the emerging concepts about non-mammalian glomeruli.


Subject(s)
Carps/physiology , Olfactory Bulb/metabolism , Olfactory Pathways/physiology , Olfactory Receptor Neurons/drug effects , Smell , Animals , Coloring Agents , Dextrans , Hypoxanthines/pharmacology , Olfactory Bulb/drug effects , Olfactory Pathways/drug effects , Olfactory Receptor Neurons/metabolism , Synapses/metabolism , Taurolithocholic Acid/pharmacology
9.
Arch Toxicol ; 89(6): 979-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-24912783

ABSTRACT

At present, it has not been systematically evaluated whether the functional alterations induced by cholestatic compounds in canalicular transporters involved in bile formation can be reproduced in sandwich-cultured rat hepatocytes (SCRHs). Here, we focused on two clinically relevant cholestatic agents, such as estradiol 17ß-D-glucuronide (E17G) and taurolithocholate (TLC), also testing the ability of dibutyryl cyclic AMP (DBcAMP) to prevent their effects. SCRHs were incubated with E17G (200 µM) or TLC (2.5 µM) for 30 min, with or without pre-incubation with DBcAMP (10 µM) for 15 min. Then, the increase in glutathione methyl fluorescein (GS-MF)-associated fluorescence inside the canaliculi was monitored by quantitative time-lapse imaging, and Mrp2 transport activity was calculated by measuring the slope of the time-course fluorescence curves during the initial linear phase, which was considered to be the Mrp2-mediated initial transport rate (ITR). E17G and TLC impaired canalicular bile formation, as evidenced by a decrease in both the bile canaliculus volume and the bile canaliculus width, estimated from 3D and 2D confocal images, respectively. These compounds decreased ITR and induced retrieval of Mrp2, a main pathomechanism involved in their cholestatic effects. Finally, DBcAMP prevented these effects, and its well-known choleretic effect was evident from the increase in the canalicular volume/width values; this choleretic effect is associated in part with its capability to increase Mrp2 activity, evidenced here by the increase in ITR of GS-MF. Our study supports the use of SCRHs as an in vitro model useful to quantify canalicular transport function under conditions of cholestasis and choleresis.


Subject(s)
Bile Canaliculi/metabolism , Bile/metabolism , Biological Transport , Cholestasis/metabolism , Hepatocytes/metabolism , Models, Biological , Animals , Bile Canaliculi/drug effects , Bucladesine/pharmacology , Cell Culture Techniques , Cells, Cultured , Cholestasis/chemically induced , Estradiol/analogs & derivatives , Estradiol/pharmacology , Hepatocytes/drug effects , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Rats , Taurolithocholic Acid/pharmacology
10.
J Biol Chem ; 288(29): 21065-21073, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23744075

ABSTRACT

Biliary pancreatitis is the most common etiology of acute pancreatitis, accounting for 30-60% of cases. A dominant theory for the development of biliary pancreatitis is the reflux of bile into the pancreatic duct and subsequent exposure to pancreatic acinar cells. Bile acids are known to induce aberrant Ca(2+) signals in acinar cells as well as nuclear translocation of NF-κB. In this study, we examined the role of the downstream Ca(2+) target calcineurin on NF-κB translocation. Freshly isolated mouse acinar cells were infected for 24 h with an adenovirus expressing an NF-κB luciferase reporter. The bile acid taurolithocholic acid-3-sulfate caused NF-κB activation at concentrations (500 µm) that were associated with cell injury. We show that the NF-κB inhibitor Bay 11-7082 (1 µm) blocked translocation and injury. Pretreatment with the Ca(2+) chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, the calcineurin inhibitors FK506 and cyclosporine A, or use of acinar cells from calcineurin Aß-deficient mice each led to reduced NF-κB activation with taurolithocholic acid-3-sulfate. Importantly, these manipulations did not affect LPS-induced NF-κB activation. A critical upstream regulator of NF-κB activation is protein kinase C, which translocates to the membranes of various organelles in the active state. We demonstrate that pharmacologic and genetic inhibition of calcineurin blocks translocation of the PKC-δ isoform. In summary, bile-induced NF-κB activation and acinar cell injury are mediated by calcineurin, and a mechanism for this important early inflammatory response appears to be upstream at the level of PKC translocation.


Subject(s)
Acinar Cells/metabolism , Bile Acids and Salts/pharmacology , Calcineurin/metabolism , NF-kappa B/metabolism , Pancreas/pathology , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Models, Biological , Protein Kinase C-delta/metabolism , Protein Transport/drug effects , Rats , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
11.
Am J Physiol Gastrointest Liver Physiol ; 307(5): G574-81, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25012845

ABSTRACT

Physiological calcium (Ca(2+)) signals within the pancreatic acinar cell regulate enzyme secretion, whereas aberrant Ca(2+) signals are associated with acinar cell injury. We have previously identified the ryanodine receptor (RyR), a Ca(2+) release channel on the endoplasmic reticulum, as a modulator of these pathological signals. In the present study, we establish that the RyR is expressed in human acinar cells and mediates acinar cell injury. We obtained pancreatic tissue from cadaveric donors and identified isoforms of RyR1 and RyR2 by qPCR. Immunofluorescence staining of the pancreas showed that the RyR is localized to the basal region of the acinar cell. Furthermore, the presence of RyR was confirmed from isolated human acinar cells by tritiated ryanodine binding. To determine whether the RyR is functionally active, mouse or human acinar cells were loaded with the high-affinity Ca(2+) dye (Fluo-4 AM) and stimulated with taurolithocholic acid 3-sulfate (TLCS) (500 µM) or carbachol (1 mM). Ryanodine (100 µM) pretreatment reduced the magnitude of the Ca(2+) signal and the area under the curve. To determine the effect of RyR blockade on injury, human acinar cells were stimulated with pathological stimuli, the bile acid TLCS (500 µM) or the muscarinic agonist carbachol (1 mM) in the presence or absence of the RyR inhibitor ryanodine. Ryanodine (100 µM) caused an 81% and 47% reduction in acinar cell injury, respectively, as measured by lactate dehydrogenase leakage (P < 0.05). Taken together, these data establish that the RyR is expressed in human acinar cells and that it modulates acinar Ca(2+) signals and cell injury.


Subject(s)
Acinar Cells/metabolism , Pancreas/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Acinar Cells/drug effects , Animals , Calcium/metabolism , Carbachol/pharmacology , Cell Death , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Pancreas/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
12.
Arterioscler Thromb Vasc Biol ; 33(7): 1663-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23619297

ABSTRACT

OBJECTIVE: TGR5 is a G-protein-coupled receptor for bile acids. So far, little is known about the function of TGR5 in vascular endothelial cells. APPROACH AND RESULTS: In bovine aortic endothelial cells, treatment with a bile acid having a high affinity to TGR5, taurolithocholic acid (TLCA), significantly increased NO production. This effect was abolished by small interfering RNA-mediated depletion of TGR5. TLCA-induced NO production was also observed in human umbilical vein endothelial cells measured via intracellular cGMP accumulation. TLCA increased endothelial NO synthase(ser1177) phosphorylation in human umbilical vein endothelial cells. This response was accompanied by increased Akt(ser473) phosphorylation and intracellular Ca(2+). Inhibition of these signals significantly decreased TLCA-induced NO production. We next examined whether TGR5-mediated NO production affects inflammatory responses of endothelial cells. In human umbilical vein endothelial cells, TLCA significantly reduced tumor necrosis factor-α-induced adhesion of monocytes, vascular cell adhesion molecule-1 expression, and activation of nuclear factor-κB. TLCA also inhibited lipopolysaccharide-induced monocyte adhesion to mesenteric venules in vivo. These inhibitory effects of TLCA were abrogated by NO synthase inhibition. CONCLUSIONS: TGR5 agonism induces NO production via Akt activation and intracellular Ca(2+) increase in vascular endothelial cells, and this function inhibits monocyte adhesion in response to inflammatory stimuli.


Subject(s)
Cell Adhesion/drug effects , Endothelial Cells/drug effects , Monocytes/drug effects , Nitric Oxide/metabolism , Receptors, G-Protein-Coupled/agonists , Taurolithocholic Acid/pharmacology , Animals , Calcium Signaling/drug effects , Cattle , Coculture Techniques , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , U937 Cells , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
13.
Nat Microbiol ; 9(10): 2583-2599, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39294459

ABSTRACT

Bile acids are microbial metabolites that can impact infection of enteric and hepatitis viruses, but their functions during systemic viral infection remain unclear. Here we show that elevated levels of the secondary bile acid taurolithocholic acid (TLCA) are associated with reduced fatality rates and suppressed viraemia in patients infected with severe fever with thrombocytopenia syndrome virus (SFTSV), an emerging tick-borne haemorrhagic fever virus. TLCA inhibits viral replication and mitigates host inflammation during SFTSV infection in vitro, and indirectly suppresses SFTSV-mediated induction of ferroptosis by upregulating fatty acid desaturase 2 via the TGR5-PI3K/AKT-SREBP2 axis. High iron and ferritin serum levels during early infection were correlated with decreased TLCA levels and fatal outcomes in SFTSV-infected patients, indicating potential biomarkers. Furthermore, treatment with either ferroptosis inhibitors or TLCA protected mice from lethal SFTSV infection. Our findings highlight the therapeutic potential of bile acids to treat haemorrhagic fever viral infection.


Subject(s)
Ferroptosis , Taurolithocholic Acid , Ferroptosis/drug effects , Animals , Mice , Humans , Taurolithocholic Acid/pharmacology , Taurolithocholic Acid/analogs & derivatives , Virus Replication/drug effects , Iron/metabolism , Male , Female , Mice, Inbred C57BL , Viremia/drug therapy , Bile Acids and Salts/metabolism
14.
J Lipid Res ; 54(12): 3334-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24115227

ABSTRACT

Increased levels of bile acids (BAs) due to the various hepatic diseases could interfere with the metabolism of xenobiotics, such as drugs, and endobiotics including steroid hormones. UDP-glucuronosyltransferases (UGTs) are involved in the conjugation and elimination of many xenobiotics and endogenous compounds. The present study sought to investigate the potential for inhibition of UGT enzymes by BAs. The results showed that taurolithocholic acid (TLCA) exhibited the strongest inhibition toward UGTs, followed by lithocholic acid. Structure-UGT inhibition relationships of BAs were examined and in vitro-in vivo extrapolation performed by using in vitro inhibition kinetic parameters (Ki) in combination with calculated in vivo levels of TLCA. Substitution of a hydrogen with a hydroxyl group in the R1, R3, R4, R5 sites of BAs significantly weakens their inhibition ability toward most UGTs. The in vivo inhibition by TLCA toward UGT forms was determined with following orders of potency: UGT1A4 > UGT2B7 > UGT1A3 > UGT1A1 ∼ UGT1A7 ∼ UGT1A10 ∼ UGT2B15. In conclusion, these studies suggest that disrupted homeostasis of BAs, notably taurolithocholic acid, found in various diseases such as cholestasis, could lead to altered metabolism of xenobiotics and endobiotics through inhibition of UGT enzymes.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Glucuronosyltransferase/metabolism , Metabolic Diseases/enzymology , Taurolithocholic Acid/pharmacology , Biocatalysis/drug effects , Glucuronosyltransferase/chemistry , Humans , Hymecromone/metabolism , Intestines/enzymology , Kinetics , Liver/enzymology , Models, Molecular , Protein Conformation , Trifluoperazine/metabolism
15.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1423-33, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22517774

ABSTRACT

Biliary pancreatitis is the most common etiology for acute pancreatitis, yet its pathophysiological mechanism remains unclear. Ca(2+) signals generated within the pancreatic acinar cell initiate the early phase of pancreatitis, and bile acids can elicit anomalous acinar cell intracellular Ca(2+) release. We previously demonstrated that Ca(2+) released via the intracellular Ca(2+) channel, the ryanodine receptor (RyR), contributes to the aberrant Ca(2+) signal. In this study, we examined whether RyR inhibition protects against pathological Ca(2+) signals, acinar cell injury, and pancreatitis from bile acid exposure. The bile acid tauro-lithocholic acid-3-sulfate (TLCS) induced intracellular Ca(2+) oscillations at 50 µM and a peak-plateau signal at 500 µM, and only the latter induced acinar cell injury, as determined by lactate dehydrogenase (LDH) leakage. Pretreatment with the RyR inhibitors dantrolene or ryanodine converted the peak-plateau signal to a mostly oscillatory pattern (P < 0.05). They also reduced acinar cell LDH leakage, basolateral blebbing, and propidium iodide uptake (P < 0.05). In vivo, a single dose of dantrolene (5 mg/kg), given either 1 h before or 2 h after intraductal TLCS infusion, reduced the severity of pancreatitis down to the level of the control (P < 0.05). These results suggest that the severity of biliary pancreatitis may be ameliorated by the clinical use of RyR inhibitors.


Subject(s)
Bile Acids and Salts/pharmacology , Calcium Signaling/physiology , Pancreatitis/etiology , Ryanodine Receptor Calcium Release Channel/metabolism , Taurolithocholic Acid/analogs & derivatives , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Calcium Signaling/drug effects , Dantrolene/pharmacology , Male , Mice , Pancreatitis/chemically induced , Pancreatitis/metabolism , Ryanodine/pharmacology , Taurolithocholic Acid/pharmacology
16.
Pancreatology ; 12(3): 248-56, 2012.
Article in English | MEDLINE | ID: mdl-22687381

ABSTRACT

BACKGROUND: Bile acids are the initiating factors of biliary acute pancreatitis. Bile acids can induce the activation of intracellular zymogen, thus leading injury in pancreatic acinar cells. Pathological zymogen activation in pancreatic acinar cells is a common feature of all types of acute pancreatitis. The proteins expressed in pancreatic acinar cells during the activation of zymogen may determine the severity of acute pancreatitis. The present study aims to determine the differentially expressed proteins in taurolithocholic acid 3-sulfate-stimulated pancreatic acinar cells as an in vitro model for acute pancreatitis. METHODS: Rat pancreatic acinar AR42J cells were treated with taurolithocholic acid 3-sulfate for 20 min. Laser confocal scanning microscopy and flow cytometry were used to detect activated trypsinogen in pancreatic acinar AR42J cells. After the determination of trypsinogen activation, proteome analysis was performed to identify the proteins differentially expressed in taurolithocholic acid 3-sulfate-treated cells and non-treated cells. RESULTS: After treatment with taurolithocholic acid 3-sulfate for 20 min, the activation of trypsinogen in AR42J cells was concurrent with changes in the protein expression profile. Thirty-nine differentially expressed proteins were detected; among these, 23 proteins were up-regulated and 16 proteins were down-regulated. KEGG analysis indicated that these proteins are involved in cellular metabolic pathways, cellular defensive mechanisms, intracellular calcium regulation and cytoskeletal changes. CONCLUSION: The expression of proteins in the pancreatic acinar cell changes at the early stage of biliary acute pancreatitis. These differentially expressed proteins will provide valuable information to understand the pathophysiologic mechanism biliary acute pancreatitis and may be useful for prognostic indices of acute pancreatitis.


Subject(s)
Acinar Cells/drug effects , Pancreatitis/physiopathology , Taurolithocholic Acid/analogs & derivatives , Acinar Cells/enzymology , Acute Disease , Animals , Cells, Cultured , Down-Regulation , Enzyme Activation , Gene Expression Profiling , Pancreas/pathology , Proteome , Rats , Taurolithocholic Acid/pharmacology , Trypsinogen/metabolism , Up-Regulation
17.
J Exp Biol ; 214(Pt 1): 80-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21147971

ABSTRACT

In the present study, we exposed the olfactory epithelia of crucian carp, Carassius carassius, and brown trout, Salmo trutta, to dextran coupled with Alexa dyes together with odorants. Dye uptake was severely reduced after pre-exposure to nocodazole, an inhibitor of microtubule polymerization that impairs endocytosis, supporting the hypothesis that odour-activated olfactory receptor molecules undergo endocytosis. Application of the bile acid taurolithocholate, a potent and specific odorant for fish, resulted in the labelling of a sparse (less than 3%) cell population with the typical morphology of ciliated sensory neurons (CSNs) - long dendrites and cell somata deep in the sensory epithelium. The dye was distributed throughout the sensory neuron, also revealing axons and target glomeruli. Stained axons redistribute at the entrance of the olfactory bulb and terminate in two small target areas, a dorsal and a medial one. These results are consistent with the notion that taurolithocholate is detected specifically by a few ciliated sensory neurons. Application of the olfactory epithelium of brown trout to bile acid stained cells with the appearance of CSNs. Application of an alarm agonist, hypxanthine-3-N-oxide, to crucian carp olfactory organ caused staining of another set of sensory neurons. Furthermore, our results show that odour-induced uptake of a dye can serve to identify the subtype of olfactory sensory neurons responding to a particular odorant, and to pinpoint the target regions of these neurons in the olfactory bulb as a first step to elucidating the neuronal network responding to a particular odour.


Subject(s)
Carps/metabolism , Nerve Net/cytology , Olfactory Receptor Neurons/drug effects , Taurolithocholic Acid/pharmacology , Trout/metabolism , Animals , Coloring Agents/pharmacokinetics , Dextrans , Endocytosis/drug effects , Image Processing, Computer-Assisted , Microscopy, Confocal , Microscopy, Electron, Scanning , Nerve Net/drug effects , Nocodazole/toxicity , Olfactory Receptor Neurons/metabolism , Staining and Labeling/methods
18.
Glia ; 58(15): 1794-805, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20665558

ABSTRACT

TGR5 (Gpbar-1) is a membrane-bound bile acid receptor in the gastrointestinal tract and immune cells with pleiotropic actions. As shown in the present study, TGR5 is also expressed in astrocytes and neurons. Here, TGR5 may act as a neurosteroid receptor, which is activated by nanomolar concentrations of 5ß-pregnan-3α-ol-20-one and micromolar concentrations of 5ß-pregnan-3α-17α-21-triol-20-one and 5α-pregnan-3α-ol-20-one (allopregnanolone). TGR5 stimulation in astrocytes and neurons is coupled to adenylate cyclase activation, elevation of intracellular Ca(2+) and the generation of reactive oxygen species. In cultured rat astrocytes, TGR5 mRNA is downregulated in the presence of neurosteroids and ammonia already at concentrations of 0.5 mmol L(-1). Furthermore, TGR5 protein levels are significantly reduced in isolated rat astrocytes after incubation with ammonia. A marked downregulation of TGR5 mRNA is also found in cerebral cortex from cirrhotic patients dying with hepatic encephalopathy (HE) when compared with brains from noncirrhotic control subjects. It is concluded that TGR5 is a novel neurosteroid receptor in brain with implications for the pathogenesis of HE.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Gene Expression Regulation/physiology , Neurons/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/cytology , CREB-Binding Protein/metabolism , Calcium/metabolism , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Coculture Techniques , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Luminescent Proteins , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurotransmitter Agents/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Statistics, Nonparametric , Taurolithocholic Acid/pharmacology , Transfection/methods
19.
ACS Infect Dis ; 6(4): 603-612, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31851822

ABSTRACT

Biofilm inhibition by exogenous molecules has been an attractive strategy for the development of novel therapeutics. We investigated the biofilm inhibitor taurolithocholic acid (TLCA) and its effects on the specialized metabolism, virulence, and biofilm formation of the clinically relevant bacterium Pseudomonas aeruginosa strain PA14. Our study shows that TLCA alters the specialized metabolism, thereby affecting P. aeruginosa colony biofilm physiology. We observed an upregulation of metabolites correlated to virulence such as the siderophore pyochelin. A wax moth virulence assay confirmed that treatment with TLCA increases the virulence of P. aeruginosa. On the basis of our results, we believe that future endeavors to identify biofilm inhibitors must consider how a putative lead alters the specialized metabolism of a bacterial community to prevent pathogens from entering a highly virulent state.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Pseudomonas aeruginosa/drug effects , Taurolithocholic Acid/pharmacology , Biofilms/growth & development , Metabolic Networks and Pathways/drug effects , Pseudomonas aeruginosa/pathogenicity , Virulence/drug effects
20.
Pancreas ; 47(1): 18-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29200128

ABSTRACT

OBJECTIVES: Mitochondrial permeability transition pore inhibition is a promising approach to treat acute pancreatitis (AP). We sought to determine (i) the effects of the mitochondrial permeability transition pore inhibitor 3,5-seco-4-nor-cholestan-5-one oxime-3-ol (TRO40303) on murine and human pancreatic acinar cell (PAC) injury induced by fatty acid ethyl esters (FAEEs) or taurolithocholic acid-3-sulfate and (ii) TRO40303 pharmacokinetics and efficacy in experimental alcoholic AP (FAEE-AP). METHODS: Changes in mitochondrial membrane potential (Δψm), cytosolic Ca ([Ca]c), and cell fate were examined in freshly isolated murine or human PACs by confocal microscopy. TRO40303 pharmacokinetics were assessed in cerulein-induced AP and therapeutic efficacy in FAEE-AP induced with palmitoleic acid and ethanol. Severity of AP was assessed by standard biomarkers and blinded histopathology. RESULTS: TRO40303 prevented loss of Δψm and necrosis induced by 100 µM palmitoleic acid ethyl ester or 500 µM taurolithocholic acid-3-sulfate in murine and human PACs. Pharmacokinetic analysis found TRO40303 accumulated in the pancreas. A single dose of 3 mg/kg TRO40303 significantly reduced serum amylase (P = 0.043), pancreatic trypsin (P = 0.018), and histopathology scores (P = 0.0058) in FAEE-AP. CONCLUSIONS: TRO40303 protects mitochondria and prevents necrotic cell death pathway activation in murine and human PACs, ameliorates the severity of FAEE-AP, and is a candidate drug for human AP.


Subject(s)
Esters/pharmacology , Fatty Acids/pharmacology , Mitochondria/drug effects , Oximes/pharmacology , Pancreatitis, Alcoholic/prevention & control , Secosteroids/pharmacology , Acinar Cells/drug effects , Acinar Cells/metabolism , Acute Disease , Animals , Ceruletide , Esters/metabolism , Fatty Acids/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/metabolism , Necrosis/prevention & control , Oximes/pharmacokinetics , Pancreatitis/chemically induced , Pancreatitis/prevention & control , Pancreatitis, Alcoholic/metabolism , Pancreatitis, Alcoholic/pathology , Secosteroids/pharmacokinetics , Taurolithocholic Acid/analogs & derivatives , Taurolithocholic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL