Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 14(12): 848, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38123563

ABSTRACT

TRIC-A and TRIC-B proteins form homotrimeric cation-permeable channels in the endoplasmic reticulum (ER) and nuclear membranes and are thought to contribute to counterionic flux coupled with store Ca2+ release in various cell types. Serious mutations in the TRIC-B (also referred to as TMEM38B) locus cause autosomal recessive osteogenesis imperfecta (OI), which is characterized by insufficient bone mineralization. We have reported that Tric-b-knockout mice can be used as an OI model; Tric-b deficiency deranges ER Ca2+ handling and thus reduces extracellular matrix (ECM) synthesis in osteoblasts, leading to poor mineralization. Here we report irregular cell death and insufficient ECM in long-bone growth plates from Tric-b-knockout embryos. In the knockout growth plate chondrocytes, excess pro-collagen fibers were occasionally accumulated in severely dilated ER elements. Of the major ER stress pathways, activated PERK/eIF2α (PKR-like ER kinase/ eukaryotic initiation factor 2α) signaling seemed to inordinately alter gene expression to induce apoptosis-related proteins including CHOP (CCAAT/enhancer binding protein homologous protein) and caspase 12 in the knockout chondrocytes. Ca2+ imaging detected aberrant Ca2+ handling in the knockout chondrocytes; ER Ca2+ release was impaired, while cytoplasmic Ca2+ level was elevated. Our observations suggest that Tric-b deficiency directs growth plate chondrocytes to pro-apoptotic states by compromising cellular Ca2+-handling and exacerbating ER stress response, leading to impaired ECM synthesis and accidental cell death.


Subject(s)
Endoplasmic Reticulum , Growth Plate , Animals , Mice , Growth Plate/metabolism , Mice, Knockout , Cell Death , Endoplasmic Reticulum/metabolism , Signal Transduction , Endoplasmic Reticulum Stress/genetics , Ion Channels/metabolism
2.
Am J Physiol Endocrinol Metab ; 324(5): E449-E460, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37074989

ABSTRACT

G protein-coupled receptor (GPR) 120 is expressed in enteroendocrine cells secreting glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide/gastric inhibitory polypeptide (GIP), and cholecystokinin (CCK). Although GPR120 signaling in adipose tissue and macrophages has been reported to ameliorate obesity and insulin resistance in a high long-chain triglyceride (LCT) diet, intestine-specific roles of GPR120 are unclear. To clarify the metabolic effect of GPR120 in the intestine, we generated intestine-specific GPR120-knockout (GPR120int-/-) mice. In comparison with floxed GPR120 (WT) mice, GPR120int-/- mice exhibited reduced GIP secretion and CCK action without change of insulin, GLP-1, or peptide YY (PYY) secretion after a single administration of LCT. Under a high-LCT diet, GPR120int-/- mice showed a mild reduction of body weight and substantial amelioration of insulin resistance and fatty liver. Moreover, liver and white adipose tissue (WAT) of GPR120int-/-mice exhibited increased Akt phosphorylation and reduced gene expression of suppressor of cytokine signaling (SOCS) 3, which inhibits insulin signaling. In addition, gene expression of inflammatory cytokines in WAT and lipogenic molecules in liver were reduced in GPR120int-/- mice. These findings suggest that inhibition of GPR120 signaling in intestine ameliorates insulin resistance and fatty liver under high-LCT diet feeding.NEW & NOTEWORTHY We generated novel intestine-specific GPR120-knockout (GPR120int-/-) mice and investigated the metabolic effect of GPR120 in the intestine. GPR120int-/- mice exhibited a reduction of GIP secretion and CCK action after a single administration of LCT. Under a high-LCT diet, GPR120int-/- mice showed mild improvement in obesity and marked amelioration of insulin resistance and hepatic steatosis. Our results indicate an important role of intestinal GPR120 on insulin resistance and hepatic steatosis.


Subject(s)
Diet, High-Fat , Intestines , Receptors, G-Protein-Coupled , Signal Transduction , Animals , Mice , Mice, Inbred C57BL , Intestines/metabolism , Insulin Resistance , Triglycerides/administration & dosage , Fatty Liver/metabolism , Mice, Knockout , Glucose/administration & dosage , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Obesity/metabolism , Corn Oil/administration & dosage
3.
Elife ; 112022 03 15.
Article in English | MEDLINE | ID: mdl-35287796

ABSTRACT

The growth plates are cartilage tissues found at both ends of developing bones, and vital proliferation and differentiation of growth plate chondrocytes are primarily responsible for bone growth. C-type natriuretic peptide (CNP) stimulates bone growth by activating natriuretic peptide receptor 2 (NPR2) which is equipped with guanylate cyclase on the cytoplasmic side, but its signaling pathway is unclear in growth plate chondrocytes. We previously reported that transient receptor potential melastatin-like 7 (TRPM7) channels mediate intermissive Ca2+ influx in growth plate chondrocytes, leading to activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) for promoting bone growth. In this report, we provide evidence from experiments using mutant mice, indicating a functional link between CNP and TRPM7 channels. Our pharmacological data suggest that CNP-evoked NPR2 activation elevates cellular cGMP content and stimulates big-conductance Ca2+-dependent K+ (BK) channels as a substrate for cGMP-dependent protein kinase (PKG). BK channel-induced hyperpolarization likely enhances the driving force of TRPM7-mediated Ca2+ entry and seems to accordingly activate CaMKII. Indeed, ex vivo organ culture analysis indicates that CNP-facilitated bone growth is abolished by chondrocyte-specific Trpm7 gene ablation. The defined CNP signaling pathway, the NPR2-PKG-BK channel-TRPM7 channel-CaMKII axis, likely pinpoints promising target proteins for developing new therapeutic treatments for divergent growth disorders.


Subject(s)
Growth Plate , TRPM Cation Channels , Animals , Bone Development , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chondrocytes , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Natriuretic Peptide, C-Type/pharmacology , TRPM Cation Channels/metabolism
4.
Front Immunol ; 13: 929690, 2022.
Article in English | MEDLINE | ID: mdl-36741381

ABSTRACT

Docosahexaenoic acid (DHA) is an omega-3 fatty acid that has a range of positive impacts on human health, including anti-inflammatory effects and inhibition of osteoclast formation via G-protein-coupled receptor 120 (GPR120). Orthodontic force was reported to induce tumor necrosis factor-α (TNF-α) expression, which activates osteoclast differentiation during orthodontic tooth movement (OTM). The aim of this study was to investigate the influence of DHA on TNF-α-induced osteoclast formation and OTM in vivo. We examined osteoclast formation and bone resorption within the calvaria of both wild-type (WT) and GPR120-deficient (GPR120-KO) mice injected with phosphate-buffered saline (PBS), TNF-α, TNF-α and DHA, or DHA. DHA inhibited TNF-α-induced osteoclast formation and bone resorption in WT mice but had no effect in GPR120-KO mice. OTM experiments were performed in mouse strains with or without regular injection of DHA, and the effects of DHA on osteoclast formation in the alveolar bones during OTM were examined. DHA also suppressed OTM in WT but not GPR120-KO mice. Our data showed that DHA suppresses TNF-α-induced osteoclastogenesis and bone resorption via GPR120. TNF-α has considerable significance in OTM, and therefore, DHA may also inhibit TNF-α-induced osteoclast formation and bone resorption in OTM.


Subject(s)
Bone Resorption , Osteoclasts , Receptors, G-Protein-Coupled , Animals , Mice , Bone Resorption/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/metabolism , Osteoclasts/metabolism , Receptors, G-Protein-Coupled/metabolism , Tooth Movement Techniques , Tumor Necrosis Factor-alpha/metabolism
5.
Biol Pharm Bull ; 44(9): 1167-1173, 2021.
Article in English | MEDLINE | ID: mdl-34471043

ABSTRACT

For pharmaceutical research focused on identifying novel drug target candidate molecules, it is essential to explore unknown biological phenomena, elucidate underlying molecular mechanisms and regulate biological processes based on these findings. Proteins expressed on the plasma membrane and endoplasmic reticulum (ER) membrane play important roles in linking extracellular environmental information to intracellular processes. Stimulating membranous proteins induces various kinds of changes in cells, such as alterations in gene expression levels and enzymatic activities. However, the physiological functions and endogenous ligands of many G-protein-coupled receptors (GPCRs) have not been determined, although GPCRs already constitute a large class of drug-target membrane proteins. Furthermore, the precise physiological roles played by many ER membrane proteins have not been elucidated to date. In this review article, I summarize the results of our recent studies, including the observations that the lipid sensor FFAR4/GPR120 controlled systemic energy homeostasis and that the ER membrane monovalent cation channel trimeric intracellular cation (TRIC)-B and the plasma membrane divalent cation channel transient receptor potential melastatin 7 (TRPM7) regulated bone formation. I further describe the therapeutic significance of these membranous protein-related biological processes.


Subject(s)
Membrane Proteins/agonists , Membrane Proteins/antagonists & inhibitors , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Energy Metabolism/drug effects , Humans , Membrane Proteins/metabolism , Molecular Targeted Therapy/methods , Osteogenesis/drug effects
6.
Cell Calcium ; 96: 102381, 2021 06.
Article in English | MEDLINE | ID: mdl-33647639

ABSTRACT

In macrophage biology, resident peritoneal macrophages (RPMs) and thioglycolate-elicited peritoneal macrophages (TGPMs) have been traditionally utilized as primary cultured models. RPMs and TGPMs exhibit distinct morphological, functional and metabolic characteristics, although it remains unclear how cellular Ca2+ handling differs between them. In our Fura-2 Ca2+ imaging, TGPMs displayed elevated resting Ca2+ levels, increased store Ca2+ contents and facilitated store-operated Ca2+ entry (SOCE) compared with RPMs. The intensified intracellular Ca2+ stores were enriched with major luminal Ca2+-binding proteins inducibly expressed in TGPMs. The elevated resting Ca2+ level was predominantly maintained by constitutive Ca2+ influx, probably through the transient receptor potential (TRP) family members TRPP2, TRPM7 and TRPA1. These TRP family channels seemed to be largely activated in a manner dependent on phospholipase C activity, and together with Orai channels, contributed to SOCE. Moreover, Ca2+-dependent K+ channels efficiently facilitated SOCE by enhancing the Ca2+ driving force in TGPMs. The consolidated cellular Ca2+ handling described may underlie the specialized cell-physiological features of TGPMs, such as vital proliferation, active migration and avid phagocytosis.


Subject(s)
Calcium/metabolism , Macrophages, Peritoneal/metabolism , Thioglycolates/pharmacology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Macrophages, Peritoneal/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL
7.
Angew Chem Int Ed Engl ; 59(47): 21210-21215, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32770565

ABSTRACT

We report herein a nonbiomimetic strategy for the total synthesis of the plicamine-type alkaloids zephycarinatines C and D. The key feature of the synthesis is a stereoselective reductive radical ipso-cyclization using visible-light-mediated photoredox catalysis. This cyclization enabled the construction of a 6,6-spirocyclic core structure through the addition of a carbon-centered radical onto the aromatic ring. Biological evaluation of zephycarinatines and their derivatives revealed that the synthetic derivative with a keto group displays moderate inhibitory activity against LPS-induced NO production. This approach could offer future opportunities to expand the chemical diversity of plicamine-type alkaloids as well as providing useful intermediates for their syntheses.

8.
Physiol Rev ; 100(1): 171-210, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31487233

ABSTRACT

Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Humans , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled/physiology
9.
Nat Commun ; 10(1): 4007, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488836

ABSTRACT

Gut microbiota mediates the effects of diet, thereby modifying host metabolism and the incidence of metabolic disorders. Increased consumption of omega-6 polyunsaturated fatty acid (PUFA) that is abundant in Western diet contributes to obesity and related diseases. Although gut-microbiota-related metabolic pathways of dietary PUFAs were recently elucidated, the effects on host physiological function remain unclear. Here, we demonstrate that gut microbiota confers host resistance to high-fat diet (HFD)-induced obesity by modulating dietary PUFAs metabolism. Supplementation of 10-hydroxy-cis-12-octadecenoic acid (HYA), an initial linoleic acid-related gut-microbial metabolite, attenuates HFD-induced obesity in mice without eliciting arachidonic acid-mediated adipose inflammation and by improving metabolic condition via free fatty acid receptors. Moreover, Lactobacillus-colonized mice show similar effects with elevated HYA levels. Our findings illustrate the interplay between gut microbiota and host energy metabolism via the metabolites of dietary omega-6-FAs thereby shedding light on the prevention and treatment of metabolic disorders by targeting gut microbial metabolites.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Fats, Unsaturated/therapeutic use , Fatty Acids, Unsaturated/pharmacology , Gastrointestinal Microbiome/drug effects , Obesity/metabolism , Adipose Tissue/pathology , Animals , Cell Line , Diet, Western , Dietary Supplements , Energy Metabolism , Fatty Acids, Omega-6/metabolism , Fatty Acids, Omega-6/therapeutic use , Fatty Acids, Unsaturated/metabolism , Gastrointestinal Microbiome/physiology , Humans , Inflammation/metabolism , Lactobacillus/metabolism , Linoleic Acid/metabolism , Metabolic Diseases/diet therapy , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice , Mice, Inbred C57BL , Models, Animal , Oleic Acids/metabolism
10.
Sci Signal ; 12(576)2019 04 09.
Article in English | MEDLINE | ID: mdl-30967513

ABSTRACT

During endochondral ossification of long bones, the proliferation and differentiation of chondrocytes cause them to be arranged into layered structures constituting the epiphyseal growth plate, where they secrete the cartilage matrix that is subsequently converted into trabecular bone. Ca2+ signaling has been implicated in chondrogenesis in vitro. Through fluorometric imaging of bone slices from embryonic mice, we demonstrated that live growth plate chondrocytes generated small, cell-autonomous Ca2+ fluctuations that were associated with weak and intermittent Ca2+ influx. Several genes encoding Ca2+-permeable channels were expressed in growth plate chondrocytes, but only pharmacological inhibitors of transient receptor potential cation channel subfamily M member 7 (TRPM7) reduced the spontaneous Ca2+ fluctuations. The TRPM7-mediated Ca2+ influx was likely activated downstream of basal phospholipase C activity and was potentiated upon cell hyperpolarization induced by big-conductance Ca2+-dependent K+ channels. Bones from embryos in which Trpm7 was conditionally knocked out during ex vivo culture exhibited reduced outgrowth and displayed histological abnormalities accompanied by insufficient autophosphorylation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) in the growth plate. The link between TRPM7-mediated Ca2+ fluctuations and CaMKII-dependent chondrogenesis was further supported by experiments with chondrocyte-specific Trpm7 knockout mice. Thus, growth plate chondrocytes generate spontaneous, TRPM7-mediated Ca2+ fluctuations that promote self-maturation and bone development.


Subject(s)
Bone Development , Calcium Signaling , Chondrocytes/metabolism , Growth Plate/metabolism , TRPM Cation Channels/metabolism , Animals , Chondrocytes/cytology , Growth Plate/cytology , Mice
11.
Article in English | MEDLINE | ID: mdl-30949128

ABSTRACT

Docosahexaenoic acid (DHA) is an n-3 fatty acid that is an important structural component of the cell membrane. DHA exerts potent anti-inflammatory effects through G protein-coupled receptor 120 (GPR120), which is a functional receptor for n-3 fatty acids. DHA also regulates osteoclast formation and function. However, no studies have investigated the effect of DHA on inflammation-induced osteoclast formation in vivo. In the present study, we investigated whether DHA influences osteoclast formation, bone resorption and the expression of osteoclast-associated cytokines during lipopolysaccharide (LPS)-induced inflammation in vivo, and then we elucidated the underlying mechanisms by using in vitro experiments. In vitro experiments revealed both receptor activator of NF-kB ligand (RANKL)- and tumor necrosis factor-α (TNF-α)-induced osteoclast formation was inhibited by DHA. Supracalvarial administration of LPS with or without DHA was carried out for 5 days and then the number of osteoclasts, ratio of bone resorption pits and the level of type I collagen C-terminal cross-linked telopeptide were measured. All measurements were significantly lower in LPS+DHA-co-administered mice than LPS-administered mice. However, this DHA-induced inhibition was not observed in LPS-, DHA-, and selective GPR120 antagonist AH7614-co-administered mice. Furthermore, the expression of RANKL and TNF-α mRNAs was lower in the LPS+DHA-co-administered group than in the LPS-administered group in vivo. TNF-α mRNA levels were decreased in macrophages co-treated with LPS+DHA compared with cells treated with LPS in vitro. In contrast, RANKL mRNA expression levels from osteoblasts co-treated with DHA and LPS in vitro were equal to that in cells treated with LPS alone. Finally, the inhibitory effects of DHA on osteoclast formation in vitro were not observed by using osteoclast precursors from GPR120-deficient mice, and inhibition of LPS-induced osteoclast formation and bone resorption by DHA in vivo was not observed in GPR120-deficient mice. These results suggest that DHA inhibits LPS-induced osteoclast formation and bone resorption in vivo via GPR120 by inhibiting LPS-induced TNF-α production in macrophages along with direct inhibition of osteoclast formation.

12.
Nat Neurosci ; 21(6): 820-833, 2018 06.
Article in English | MEDLINE | ID: mdl-29713080

ABSTRACT

Chronic morphine exposure upregulates adenylate cyclase signaling and reduces analgesic efficacy, a condition known as opioid tolerance. Nonopioid neurotransmitters can enhance morphine tolerance, but the mechanism for this is poorly understood. We show that morphine tolerance was delayed in mice lacking vasopressin 1b receptors (V1bRs) or after administration of V1bR antagonist into the rostral ventromedial medulla, where transcripts for V1bRs and µ-opioid receptors are co-localized. Vasopressin increased morphine-binding affinity in cells expressing both V1bR and µ-opioid receptors. Complex formation among V1bR, ß-arrestin-2, and µ-opioid receptor resulted in vasopressin-mediated upregulation of ERK phosphorylation and adenylate cyclase sensitization. A leucine-rich segment in the V1bR C-terminus was necessary for the association with ß-arrestin-2. Deletion of this leucine-rich segment increased morphine analgesia and reduced vasopressin-mediated adenylate cyclase sensitization. These findings indicate that inhibition of µ-opioid-receptor-associated V1bR provides an approach for enhancing morphine analgesia without increasing analgesic tolerance.


Subject(s)
Drug Tolerance/genetics , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/metabolism , Receptors, Vasopressin/metabolism , beta-Arrestin 2/metabolism , Adenylyl Cyclases/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/genetics , Injections , MAP Kinase Signaling System/drug effects , Male , Medulla Oblongata , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacokinetics , Morphine Dependence/psychology , Narcotics/pharmacokinetics , Pain Measurement/drug effects , Pain Threshold/drug effects , Phosphorylation , Receptors, Opioid, mu/genetics , Receptors, Vasopressin/genetics , beta-Arrestin 2/genetics
13.
Molecules ; 23(4)2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29641495

ABSTRACT

Induction of a series of anti-hypoxic proteins protects cells during exposure to hypoxic conditions. Hypoxia-inducible factor-α (HIF-α) is a major transcription factor that orchestrates this protective effect. To activate HIF exogenously, without exposing cells to hypoxic conditions, many small-molecule inhibitors targeting prolyl hydroxylase domain-containing protein have been developed. In addition, suppression of factor inhibiting HIF-1 (FIH-1) has also been shown to have the potential to activate HIF-α. However, few small-molecule inhibitors of FIH-1 have been developed. In this study, we synthesized a series of furan- and thiophene-2-carbonyl amino acid derivatives having the potential to inhibit FIH-1. The inhibitory activities of these compounds were evaluated in SK-N-BE(2)c cells by measuring HIF response element (HRE) promoter activity. Several furan- and thiophene-2-carbonyl amino acid derivatives inhibited FIH-1 based on correlations among the docking score of the FIH-1 active site, the chemical structure of the compounds, and biological HIF-α/HRE transcriptional activity.


Subject(s)
Furans/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mixed Function Oxygenases/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Thiophenes/pharmacology , Catalytic Domain/drug effects , Cell Line , Furans/chemical synthesis , Furans/chemistry , Gene Expression Regulation/drug effects , Humans , Mixed Function Oxygenases/chemistry , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Repressor Proteins/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Thiophenes/chemical synthesis , Thiophenes/chemistry , Transcriptional Activation/drug effects
14.
Eur J Pharmacol ; 820: 217-221, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29274333

ABSTRACT

Myricetin (3,3',4',5,5',7-hexahydroxyflavone), a major flavonoid in berries and red wine, has been recently used as a health food supplement based on its antioxidant and antitumor properties. We report here that myricetin preferentially exerts inhibitory effects on gastric H+, K+-ATPase. Myricetin inhibited H+, K+-ATPase with a sub-micromolar IC50 value in an enzyme assay using freeze-dried tubulovesicles prepared from hog stomach. Na+, K+-ATPase and Ca2+-ATPase were also inhibited by myricetin in a dose-dependent manner, but the IC50 values for these enzymes were approximately an order of magnitude higher compared to the H+, K+-ATPase. In structure-inhibitory functional analysis of sixteen myricetin derivatives, several phenolic hydroxy groups attached to the flavonoid backbone were highlighted as essential modifications for the inhibition of P2-type ATPases. Furthermore, oral administration of myricetin significantly attenuated histamine-induced gastric acid secretion in an in vivo mouse assessment. Therefore, myricetin derivatives seem to be useful seed compounds for developing new drugs and supplements to alleviate gastric acid secretion.


Subject(s)
Biological Products/pharmacology , Flavonoids/pharmacology , H(+)-K(+)-Exchanging ATPase/metabolism , Proton Pump Inhibitors/pharmacology , Stomach/enzymology , Animals , Biological Products/chemistry , Calcium/metabolism , Flavonoids/chemistry , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Proton Pump Inhibitors/chemistry , Proton Pumps/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
15.
Biomed Res ; 38(3): 215-219, 2017.
Article in English | MEDLINE | ID: mdl-28637957

ABSTRACT

The Gm7325 gene, bioinformatically identified in the mouse genome, encodes a small protein but has not been characterized until recently. Our gene expression analysis revealed that Gm7325 transcription is remarkably upregulated in injured skeletal muscle tissues. Activated satellite cells and immature myotubes were densely decorated with positive signals for Gm7325 mRNA in in situ hybridization analysis, while no obvious signals were observed in quiescent satellite cells and mature myofibers. In the 5'-flanking regions of mouse Gm7325 and its human homologue, conserved E-box motifs for helix-loop-helix transcription factors are repeatedly arranged around the putative promoter regions. Reporter gene assays suggested that MyoD, a master transcription factor for myogenesis, binds to the conserved E-box motifs to activate Gm7325 expression. Therefore, Gm7325, as a novel MyoD-target gene, is specifically induced in activated satellite cells, and may have an important role in skeletal myogenesis.


Subject(s)
Membrane Proteins/physiology , MyoD Protein/physiology , Satellite Cells, Skeletal Muscle/physiology , Animals , Base Sequence , Cell Line , Consensus Sequence , Gene Expression , HeLa Cells , Humans , Mice , Muscle Development , Transcriptional Activation
16.
FASEB J ; 31(6): 2625-2637, 2017 06.
Article in English | MEDLINE | ID: mdl-28270519

ABSTRACT

Adhesive small bowel obstruction remains a common problem for surgeons. After surgery, platelet aggregation contributes to coagulation cascade and fibrin clot formation. With clotting, fibrin degradation is simultaneously enhanced, driven by tissue plasminogen activator-mediated cleavage of plasminogen to form plasmin. The aim of this study was to investigate the cellular events and proteolytic responses that surround plasminogen activator inhibitor (PAI-1; Serpine1) inhibition of postoperative adhesion. Peritoneal adhesion was induced by gauze deposition in the abdominal cavity in C57BL/6 mice and those that were deficient in fibrinolytic factors, such as Plat-/- and Serpine1-/- In addition, C57BL/6 mice were treated with the novel PAI-1 inhibitor, TM5275. Some animals were treated with clodronate to deplete macrophages. Epidermal growth factor (EGF) experiments were performed to understand the role of macrophages and how EGF contributes to adhesion. In the early phase of adhesive small bowel obstruction, increased PAI-1 activity was observed in the peritoneal cavity. Genetic and pharmacologic PAI-1 inhibition prevented progression of adhesion and increased circulating plasmin. Whereas Serpine1-/- mice showed intra-abdominal bleeding, mice that were treated with TM5275 did not. Mechanistically, PAI-1, in combination with tissue plasminogen activator, served as a chemoattractant for macrophages that, in turn, secreted EGF and up-regulated the receptor, HER1, on peritoneal mesothelial cells, which led to PAI-1 secretion, further fueling the vicious cycle of impaired fibrinolysis at the adhesive site. Controlled inhibition of PAI-1 not only enhanced activation of the fibrinolytic system, but also prevented recruitment of EGF-secreting macrophages. Pharmacologic PAI-1 inhibition ameliorated adhesion formation in a macrophage-dependent manner.-Honjo, K., Munakata, S., Tashiro, Y., Salama, Y., Shimazu, H., Eiamboonsert, S., Dhahri, D., Ichimura, A., Dan, T., Miyata, T., Takeda, K., Sakamoto, K., Hattori, K., Heissig, B. Plasminogen activator inhibitor-1 regulates macrophage-dependent postoperative adhesion by enhancing EGF-HER1 signaling in mice.


Subject(s)
ErbB Receptors/metabolism , Macrophages/physiology , Piperazines/therapeutic use , Serpin E2/antagonists & inhibitors , Tissue Adhesions/pathology , para-Aminobenzoates/therapeutic use , Animals , CD11b Antigen , Cell Migration Assays , Cell Movement/drug effects , Cetuximab/pharmacology , Epidermal Growth Factor , ErbB Receptors/genetics , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Postoperative Complications/prevention & control , RAW 264.7 Cells , Serpin E2/genetics , Serpin E2/metabolism , Signal Transduction , Tissue Adhesions/metabolism , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
17.
Handb Exp Pharmacol ; 236: 133-158, 2017.
Article in English | MEDLINE | ID: mdl-27864678

ABSTRACT

Overfeeding of fat can cause various metabolic disorders including obesity and type 2 diabetes (T2D). Diet provided free fatty acids (FFAs) are not only essential nutrients, but they are also recognized as signaling molecules, which stimulate various important biological functions. Recently, several G protein-coupled receptors (GPCRs), including FFA1-4, have been identified as receptors of FFAs by various physiological and pharmacological studies. FFAs exert physiological functions through these FFA receptors (FFARs) depending on carbon chain length and degree of unsaturation. Functional analyses have revealed that several important metabolic processes, such as peptide hormone secretion, cell maturation and nerve activities, are regulated by FFARs and thereby FFARs contribute to the energy homeostasis through these physiological functions. Hence, FFARs are expected to be promising pharmacological targets for metabolic disorders since imbalances in energy homeostasis lead to metabolic disorders. In human, it is established that different responses of individuals to endogenous ligands and chemical drugs may be due to differences in the ability of such ligands to activate nucleotide polymorphic variants of receptors. However, the clear links between genetic variations that are involved in metabolic disorders and polymorphisms receptors have been relatively difficult to assess. In this review, I summarize current literature describing physiological functions of FFARs and genetic variations of those receptors to discuss the potential of FFARs as drug targets for metabolic disorders.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Energy Metabolism , Genetic Variation , Humans , Mice , Polymorphism, Single Nucleotide , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics
18.
Biol Pharm Bull ; 39(11): 1743-1747, 2016.
Article in English | MEDLINE | ID: mdl-27803445

ABSTRACT

Trimeric intracellular cation (TRIC) channel subtypes, namely TRIC-A and TRIC-B, are expressed in the endoplasmic/sarcoplasmic reticulum and nuclear envelope, and likely function as monovalent cation channels in various cell types. Our studies using knockout mice so far suggest that TRIC subtypes support Ca2+ release from intracellular stores by mediating counter-cationic fluxes. Several genetic mutations within the TRIC-B locus were recently identified in autosomal recessive osteogenesis imperfecta (OI) patients. However, the molecular mechanism by which the mutations cause human disease is not fully addressed. We found that Tric-b-knockout mice exhibit poor bone ossification and thus serve as an OI-model animal. Studies on Tric-b-knockout bones and cultured cell lines derived from the patients currently reveal the main part of the pathophysiological mechanism involved in the TRIC-B-mutated OI form. This mini-review focuses on the essential role of TRIC-B channels in bone ossification.


Subject(s)
Ion Channels/genetics , Osteogenesis Imperfecta/genetics , Animals , Humans , Mutation
19.
Sci Signal ; 9(428): ra49, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27188440

ABSTRACT

The trimeric intracellular cation (TRIC) channels TRIC-A and TRIC-B localize predominantly to the endoplasmic reticulum (ER) and likely support Ca(2+) release from intracellular stores by mediating cationic flux to maintain electrical neutrality. Deletion and point mutations in TRIC-B occur in families with autosomal recessive osteogenesis imperfecta. Tric-b knockout mice develop neonatal respiratory failure and exhibit poor bone ossification. We investigated the cellular defect causing the bone phenotype. Bone histology indicated collagen matrix deposition was reduced in Tric-b knockout mice. Osteoblasts, the bone-depositing cells, from Tric-b knockout mice exhibited reduced Ca(2+) release from ER and increased ER Ca(2+) content, which was associated with ER swelling. These cells also had impaired collagen release without a decrease in collagen-encoding transcripts, consistent with a defect in trafficking of collagen through ER. In contrast, osteoclasts, the bone-degrading cells, from Tric-b knockout mice were similar to those from wild-type mice. Thus, TRIC-B function is essential to support the production and release of large amounts of collagen by osteoblasts, which is necessary for bone mineralization.


Subject(s)
Bone and Bones/metabolism , Calcification, Physiologic , Collagen/metabolism , Ion Channels/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Cations/metabolism , Collagen/chemistry , Endoplasmic Reticulum/metabolism , Female , Femur/metabolism , Homeostasis , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Skull/metabolism , X-Ray Microtomography
20.
Int J Mol Sci ; 17(4): 450, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-27023530

ABSTRACT

Excess energy is stored primarily as triglycerides, which are mobilized when demand for energy arises. Dysfunction of energy balance by excess food intake leads to metabolic diseases, such as obesity and diabetes. Free fatty acids (FFAs) provided by dietary fat are not only important nutrients, but also contribute key physiological functions via FFA receptor (FFAR)-mediated signaling molecules, which depend on FFAs' carbon chain length and the ligand specificity of the receptors. Functional analyses have revealed that FFARs are critical for metabolic functions, such as peptide hormone secretion and inflammation, and contribute to energy homeostasis. In particular, recent studies have shown that the administration of selective agonists of G protein-coupled receptor (GPR) 40 and GPR120 improved glucose metabolism and systemic metabolic disorders. Furthermore, the anti-inflammation and energy metabolism effects of short chain FAs have been linked to the activation of GPR41 and GPR43. In this review, we summarize recent progress in research on FFAs and their physiological roles in the regulation of energy metabolism.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Fatty Acids, Nonesterified/metabolism , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Receptors, Cell Surface/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...