Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Int J Med Sci ; 21(8): 1461-1471, 2024.
Article in English | MEDLINE | ID: mdl-38903922

ABSTRACT

Dasatinib is one of the second-generation tyrosine kinase inhibitors used to treat chronic myeloid leukemia and has a broad target spectrum, including KIT, PDGFR, and SRC family kinases. Due to its broad drug spectrum, dasatinib has been reported at the basic research level to improve athletic performance by eliminating senescent cell removal and to have an effect on muscle diseases such as Duchenne muscular dystrophy, but its effect on myoblasts has not been investigated. In this study, we evaluated the effects of dasatinib on skeletal muscle both under normal conditions and in the regenerating state. Dasatinib suppressed the proliferation and promoted the fusion of C2C12 myoblasts. During muscle regeneration, dasatinib increased the gene expressions of myogenic-related genes (Myod, Myog, and Mymx), and caused abnormally thin muscle fibers on the CTX-induced muscle injury mouse model. From these results, dasatinib changes the closely regulated gene expression pattern of myogenic regulatory factors during muscle differentiation and disrupts normal muscle regeneration. Our data suggest that when using dasatinib, its effects on skeletal muscle should be considered, particularly at regenerating stages.


Subject(s)
Cell Differentiation , Dasatinib , Muscle Development , Muscle, Skeletal , Myoblasts , Regeneration , Dasatinib/pharmacology , Animals , Mice , Regeneration/drug effects , Cell Differentiation/drug effects , Muscle Development/drug effects , Muscle Development/genetics , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/cytology , Cell Proliferation/drug effects , Humans , Cell Line , Protein Kinase Inhibitors/pharmacology
2.
Hepatol Commun ; 8(6)2024 06 01.
Article in English | MEDLINE | ID: mdl-38836815

ABSTRACT

BACKGROUND: Smoking is a risk factor for liver cirrhosis; however, the underlying mechanisms remain largely unexplored. The α7 nicotinic acetylcholine receptor (α7nAChR) has recently been detected in nonimmune cells possessing immunoregulatory functions. We aimed to verify whether nicotine promotes liver fibrosis via α7nAChR. METHODS: We used osmotic pumps to administer nicotine and carbon tetrachloride to induce liver fibrosis in wild-type and α7nAChR-deficient mice. The severity of fibrosis was evaluated using Masson trichrome staining, hydroxyproline assays, and real-time PCR for profibrotic genes. Furthermore, we evaluated the cell proliferative capacity and COL1A1 mRNA expression in human HSCs line LX-2 and primary rat HSCs treated with nicotine and an α7nAChR antagonist, methyllycaconitine citrate. RESULTS: Nicotine exacerbated carbon tetrachloride-induced liver fibrosis in mice (+42.4% in hydroxyproline assay). This effect of nicotine was abolished in α7nAChR-deficient mice, indicating nicotine promotes liver fibrosis via α7nAChR. To confirm the direct involvement of α7nAChRs in liver fibrosis, we investigated the effects of genetic suppression of α7nAChR expression on carbon tetrachloride-induced liver fibrosis without nicotine treatment. Profibrotic gene expression at 1.5 weeks was significantly suppressed in α7nAChR-deficient mice (-83.8% in Acta2, -80.6% in Col1a1, -66.8% in Tgfb1), and collagen content was decreased at 4 weeks (-22.3% in hydroxyproline assay). The in vitro analysis showed α7nAChR expression in activated but not in quiescent HSCs. Treatment of LX-2 cells with nicotine increased COL1A1 expression (+116%) and cell proliferation (+10.9%). These effects were attenuated by methyllycaconitine citrate, indicating the profibrotic effects of nicotine via α7nAChR. CONCLUSIONS: Nicotine aggravates liver fibrosis induced by other factors by activating α7nAChR on HSCs, thereby increasing their collagen-producing capacity. We suggest the profibrotic effect of nicotine is mediated through α7nAChRs.


Subject(s)
Collagen Type I, alpha 1 Chain , Collagen Type I , Hepatic Stellate Cells , Liver Cirrhosis , Nicotine , alpha7 Nicotinic Acetylcholine Receptor , Animals , Humans , Mice , Rats , Aconitine/pharmacology , Aconitine/analogs & derivatives , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Carbon Tetrachloride/toxicity , Cell Line , Cell Proliferation/drug effects , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Mice, Knockout , Nicotine/adverse effects , Transforming Growth Factor beta1/metabolism
3.
J Vet Med Sci ; 84(4): 610-617, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35249909

ABSTRACT

Postoperative ileus (POI) is a postsurgical gastrointestinal motility dysfunction caused by mechanical stress to the intestine during abdominal surgery. POI leads to nausea and vomiting reduced patient quality of life, as well as high medical costs and extended hospitalization. Intestinal inflammation caused by macrophages and neutrophils is thought to be important in the mechanism of POI. Surgery-associated tissue injury and inflammation induce the release of adenosine triphosphate (ATP) from injured cells. Released ATP binds the purinergic P2X7 receptor (P2X7R) expressed on inflammatory cells, inducing the secretion of inflammatory mediators. P2X7R antagonists are thought to be important mediators of the first step in the inflammation process, and studies in chemically induced colitis models confirmed that P2X7R antagonists exhibit anti-inflammatory effects. Therefore, we hypothesized that P2X7R plays an important role in POI. POI models were generated from C57BL/6J mice. Mice were treated with P2X7R antagonist A438079 (34 mg/kg) 30 min before and 2 hr after intestinal manipulation (IM). Inflammatory cell infiltration and gastrointestinal transit were measured. A438079 ameliorated macrophage and neutrophil infiltration in the POI model. Impaired intestinal transit improved following A438079 treatment. P2X7R was expressed on both infiltrating and resident macrophages in the inflamed ileal muscle layer. The P2X7R antagonist A438079 exhibits anti-inflammatory effects via P2X7R expressed on macrophages and therefore could be a target in the treatment of POI.


Subject(s)
Ileus , Rodent Diseases , Adenosine Triphosphate , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Ileus/drug therapy , Ileus/etiology , Ileus/metabolism , Ileus/veterinary , Inflammation/drug therapy , Inflammation/veterinary , Mice , Mice, Inbred C57BL , Postoperative Complications/drug therapy , Postoperative Complications/veterinary , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/therapeutic use , Quality of Life , Receptors, Purinergic P2X7/therapeutic use
4.
BMC Res Notes ; 14(1): 438, 2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34838130

ABSTRACT

OBJECTIVE: The biological importance for the signaling of C-type natriuretic peptide (CNP) and natriuretic peptide receptor B (NPR-B) has been recognized. However, the details remain unclear and are debatable. The Npr2 is a gene of NPR-B, and we previously reported a unique phenotype of a spontaneous mutant mouse lacking Npr2 (Npr2slw/slw), such as severe ileus-like disorder with bloodless blood vessels. In this study, we analyzed the bloodless mesenteric vascular morphology of Npr2slw/slw by histological observation to clarify the effects of the CNP/NPR-B signal deficiency. RESULTS: Blood vessels in the mesentery were clearly dilated in the preweaning Npr2slw/slw mice. Additionally, in the Npr2slw/slw mice, the lacteals were partially dilation or randomly direction mucosal epithelial cells in villi, and mesenteric adipocytes were undeveloped. These findings provide important information for understanding the role of CNP/NPR-B signals on intestine with mesentery.


Subject(s)
Natriuretic Peptide, C-Type , Vasodilation , Adipocytes , Animals , Intestines , Mesentery , Mice , Natriuretic Peptide, C-Type/genetics
5.
Cell Death Dis ; 12(1): 11, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33414474

ABSTRACT

Liver cirrhosis is a critical health problem associated with several complications, including skeletal muscle atrophy, which adversely affects the clinical outcome of patients independent of their liver functions. However, the precise mechanism underlying liver cirrhosis-induced muscle atrophy has not been elucidated. Here we show that serum factor induced by liver fibrosis leads to skeletal muscle atrophy. Using bile duct ligation (BDL) model of liver injury, we induced liver fibrosis in mice and observed subsequent muscle atrophy and weakness. We developed culture system of human primary myotubes that enables an evaluation of the effects of soluble factors on muscle atrophy and found that serum from BDL mice contains atrophy-inducing factors. This atrophy-inducing effect of BDL mouse serum was mitigated upon inhibition of TNFα signalling but not inhibition of myostatin/activin signalling. The BDL mice exhibited significantly up-regulated serum levels of TNFα when compared with the control mice. Furthermore, the mRNA expression levels of Tnf were markedly up-regulated in the fibrotic liver but not in the skeletal muscles of BDL mice. The gene expression analysis of isolated nuclei revealed that Tnf is exclusively expressed in the non-fibrogenic diploid cell population of the fibrotic liver. These findings reveal the mechanism through which circulating TNFα produced in the damaged liver mediates skeletal muscle atrophy. Additionally, this study demonstrated the importance of inter-organ communication that underlies the pathogenesis of liver cirrhosis.


Subject(s)
Liver Cirrhosis/pathology , Muscular Atrophy/etiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Humans , Male , Mice , Muscular Atrophy/pathology
6.
Sci Rep ; 9(1): 5887, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30971711

ABSTRACT

Gastrointestinal prokinetic agents function as serotonin-4 receptor (5-HT4R) agonists to activate myenteric plexus neurons to release acetylcholine (ACh), which then induce anti-inflammatory action. Details of this pathway, however, remain unknown. The aim of this study is to clarify the anti-inflammatory mechanism underlying the 5-HT4R agonist, mosapride citrate (MOS)-induced anti-inflammatory action on postoperative ileus (POI). POI models were generated from wild-type C57BL6/J (WT), 5-HT4R knock-out (S4R KO), α7 nicotinic AChR KO (α7 R KO), and M2 muscarinic ACh receptor KO (M2R KO) mice. MOS attenuated leukocyte infiltration in WT. MOS-induced anti-inflammatory action was completely abolished in both S4R KO and S4R KO mice upon wild-type bone marrow transplantation. MOS-induced anti-inflammatory action against macrophage infiltration, but not neutrophil infiltration, was attenuated in α7 R KO mice. Selective α7nAChR agonists (PNU-282987 and AR-R17779) also inhibited only macrophage infiltration in POI. MOS-mediated inhibition of neutrophil infiltration was diminished by atropine, M2AChR antagonist, methoctramine, and in M2R KO mice. Stimulation with 5-HT4R inhibits leukocyte infiltration in POI, possibly through myenteric plexus activation. Released ACh inhibited macrophage and neutrophil infiltration likely by activation of α7nAChR on macrophages and M2AChR. Thus, macrophage and neutrophil recruitment into inflamed sites is regulated by different types of AChR in the small intestine.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Intestine, Small/drug effects , Receptors, Cholinergic/metabolism , Acetylcholine/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Bridged Bicyclo Compounds/pharmacology , Bridged-Ring Compounds/pharmacology , Diamines/pharmacology , Ileus/drug therapy , Ileus/pathology , Intestine, Small/metabolism , Leukocytes/cytology , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Morpholines/therapeutic use , Receptors, Cholinergic/chemistry , Receptors, Cholinergic/genetics , Receptors, Serotonin, 5-HT4/chemistry , Receptors, Serotonin, 5-HT4/genetics , Receptors, Serotonin, 5-HT4/metabolism , Spiro Compounds/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
7.
FASEB J ; 33(2): 1669-1680, 2019 02.
Article in English | MEDLINE | ID: mdl-30207796

ABSTRACT

Antagonists of the 5-hydroxytryptamine (serotonin) 3 receptor (5-HT3R) have anti-inflammatory and anti-apoptotic activities, but the detailed, underlying mechanisms are not well understood. We focused on anti-apoptotic activities via 5-HT3R signaling to clarify the underlying mechanisms. Mice were administered 5-fluorouracil (5-FU), which induced apoptosis in intestinal epithelial cells. Coadministration with 5-HT3R antagonists or agonists tended to decrease or increase the number of apoptotic cells, respectively. In serotonin 3A receptor (5-HT3AR) null (HTR3A-/-) mice, the number of apoptotic cells induced by 5-FU was decreased compared with that in wild-type (WT) mice. Bone marrow (BM) transplantation was performed to determine if BM-derived immune cells regulated 5-FU-induced apoptosis, but they were found to be unrelated to this process. Data from 5-HT3AR/enhanced green fluorescent protein reporter mice revealed that 50% of enterochromaffin (EC) cells expressed 5-HT3AR, but the number of apoptotic cells induced by 5-FU in the intestinal crypt organoids of HTR3A-/- mice was not altered compared with WT mice. In contrast, plasma 5-HT concentrations in WT mice but not in HTR3A-/- mice administered 5-FU were increased significantly. In conclusion, 5-HT3R signaling may enhance 5-HT release, possibly from EC cells intravascularly, or paracrine, resulting in increases in plasma 5-HT concentration, which in turn, enhances apoptotic activities induced by 5-FU.-Mikawa, S., Kondo, M., Kaji, N., Mihara, T., Yoshitake, R., Nakagawa, T., Takamoto, M., Nishimura, R., Shimada, S., Ozaki, H., Hori, M. Serotonin 3 receptor signaling regulates 5-fluorouracil-mediated apoptosis indirectly via TNF-α production by enhancing serotonin release from enterochromaffin cells.


Subject(s)
Antimetabolites/pharmacology , Apoptosis/drug effects , Enterochromaffin Cells/drug effects , Fluorouracil/pharmacology , Serine Endopeptidases/metabolism , Serotonin/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Bone Marrow Cells/cytology , Enterochromaffin Cells/metabolism , Green Fluorescent Proteins/genetics , Intestine, Small/cytology , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Serine Endopeptidases/genetics
8.
J Vet Med Sci ; 79(11): 1795-1802, 2017 Nov 10.
Article in English | MEDLINE | ID: mdl-28931778

ABSTRACT

Regulation of inflammation in intestinal mesothelial cells in the abdominal cavity is important for the pathogeny of clinical conditions, such as postoperative ileus, peritonitis and encapsulating peritoneal sclerosis. Here we have examined the inflammatory effect of lipopolysaccharide (LPS) and the anti-inflammatory effect of nicotinic acetylcholine receptor stimulation in rat intestinal mesothelial cells. LPS upregulated mRNA expression of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), monocyte chemotactic protein-1 (MCP-1) and inducible nitric oxide synthase (iNOS). The α7, α9 and α10 subunits of nicotinic acetylcholine receptor were detected in intestinal mesothelial cells. Nicotine (10 nM) significantly inhibited LPS-induced mRNA expression of IL-1ß and iNOS, but not TNF-α and MCP-1. In addition, the α7 nicotinic acetylcholine receptor selective agonist, PNU-282987 (10 nM), significantly inhibited LPS-induced mRNA expression of IL-1ß but not TNF-α, iNOS and MCP-1. Finally, we found that enteric nerves adhered to intestinal mesothelial cells located under the ileal serosa. In conclusion, intestinal mesothelial cells react to LPS to induce the production of nitric oxide from iNOS. The anti-inflammatory action of intestinal mesothelial cells expressing α7nAChR may be mediated via their connectivity with enteric nerves.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa/metabolism , Receptors, Nicotinic/metabolism , Animals , Inflammation/physiopathology , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Intestinal Mucosa/ultrastructure , Male , Microscopy, Electron , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/physiology , Signal Transduction/physiology
9.
Inflammation ; 40(4): 1409-1415, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28493078

ABSTRACT

The 5-hydroxytryptamine-3 receptor (5-HT3R) antagonist ondansetron has been clinically approved as an anti-emetic agent. Recent findings indicate that ondansetron has anti-inflammatory properties. The aim of the present study was to assess the therapeutic action of ondansetron in cerulein-induced acute pancreatitis model. Male-BALB/c mice were used in the present study. Acute pancreatitis was induced by an hourly injection of cerulein. Ondansetron was administered subcutaneously at a dose of 3 mg/kg. The messenger RNA (mRNA) expression of 5-HT3 R in pancreatic tissue was assessed with RT-PCR. Plasma amylase, lipase, and interleukin (IL)-6 levels were evaluated. Pancreatic injury was histopathologically graded, and myeloperoxidase (MPO)-positive cells were counted. 5-HT3R mRNA was expressed in the pancreas. In acute pancreatitis model mice, amylase, lipase, and IL-6 levels were significantly increased in the blood. With ondansetron treatment, these levels were significantly decreased. Histopathological evaluation revealed that ondansetron attenuated the inflammatory damage in acute pancreatitis. The number of infiltrated neutrophils stained by MPO was decreased by ondansetron treatment. In summary, the 5-HT3R antagonist ondansetron attenuated pancreatic injury through its anti-inflammatory action. These findings suggest that ondansetron may potentially be of use for therapy of acute pancreatitis.


Subject(s)
Ondansetron/therapeutic use , Pancreatitis/drug therapy , Acute Disease , Amylases/blood , Animals , Ceruletide/adverse effects , Interleukin-6/blood , Lipase/blood , Male , Mice , Mice, Inbred BALB C , Ondansetron/pharmacology , Pancreas/injuries , Pancreatitis/chemically induced , Serotonin 5-HT3 Receptor Antagonists/therapeutic use
10.
Inflammation ; 40(4): 1331-1341, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28493080

ABSTRACT

Postoperative ileus is a common complication after intra-abdominal surgery. Nitric oxide produced by macrophages in the inflamed gastrointestinal tract plays a crucial role in the pathogeny of postoperative ileus. Honokiol, extracted from the bark of Magnolia spp., is a natural compound with a biphenolic structure. In the present study, we examined the effect of honokiol on postoperative ileus and discussed its site of action. Postoperative ileus model mice were generated by surgical intestinal manipulation. Mice were administered honokiol (10 mg kg-1, per os) 1 h before and after intestinal manipulation. Gastrointestinal transit, leukocyte infiltration, and messenger RNA (mRNA) expression of inflammatory mediators were measured in postoperative ileus model mice with or without honokiol. We also investigated the inflammatory effect of honokiol in lipopolysaccharide-stimulated peritoneal macrophages. Gastrointestinal transit was delayed in postoperative ileus model mice and honokiol recovered the impaired transit. Honokiol significantly inhibited leukocyte infiltration and upregulation of proinflammatory cytokines (tumor necrosis factor-α, interleukin-1ß, and interleukin-6) and inducible nitric oxide synthase in the ileal muscle layer of postoperative ileus model mice. In peritoneal macrophages activated by lipopolysaccharide, honokiol significantly inhibited the upregulated mRNA expression of proinflammatory cytokines and inducible nitric oxide synthase. Honokiol significantly recovered gastrointestinal dysmotility and inhibited intestinal inflammation in postoperative ileus. Moreover, honokiol was suggested to have effects on macrophages, namely, inhibiting mRNA expression of proinflammatory cytokines and inducible nitric oxide synthase. Taken together, honokiol represents a potential novel therapeutic agent for postoperative ileus.


Subject(s)
Biphenyl Compounds/therapeutic use , Ileus/drug therapy , Lignans/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Animals , Cytokines/genetics , Down-Regulation , Gastrointestinal Motility/drug effects , Inflammation Mediators , Leukocytes/cytology , Leukocytes/metabolism , Macrophages, Peritoneal/metabolism , Mice , Nitric Oxide Synthase Type II/genetics , Postoperative Complications/drug therapy , RNA, Messenger/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL