Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G617-G626, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33533304

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin secreted from enteroendocrine preproglucagon (PPG)-expressing cells (traditionally known as L cells) in response to luminal nutrients that potentiates insulin secretion. Augmentation of endogenous GLP-1 secretion might well represent a novel therapeutic target for diabetes treatment in addition to the incretin-associated drugs currently in use. In this study, we found that PPG cells substantially express carbonic anhydrase 8 (CAR8), which has been reported to inhibit inositol 1,4,5-trisphosphate (IP3) binding to the IP3 receptor and subsequent Ca2+ efflux from the endoplasmic reticulum in neuronal cells. In vitro experiments using STC-1 cells demonstrated that Car8 knockdown increases long-chain fatty acid (LCFA)-stimulated GLP-1 secretion. This effect was reduced in the presence of phospholipase C (PLC) inhibitor; in addition, Car8 knockdown increased the intracellular Ca2+ elevation caused by α-linolenic acid, indicating that CAR8 exerts its effect on GLP-1 secretion via the PLC/IP3/Ca2+ pathway. Car8wdl null mutant mice showed significant increase in GLP-1 response to oral corn oil administration compared with that in wild-type littermates, with no significant change in intestinal GLP-1 content. These results demonstrate that CAR8 negatively regulates GLP-1 secretion from PPG cells in response to LCFAs, suggesting the possibility of augmentation of postprandial GLP-1 secretion by CAR8 inhibition.NEW & NOTEWORTHY This study focused on the physiological significance of carbonic anhydrase 8 (CAR8) in GLP-1 secretion from enteroendocrine preproglucagon (PPG)-expressing cells. We found an inhibitory role of CAR8 in LCFA-induced GLP-1 secretion in vitro and in vivo, suggesting a novel therapeutic approach to diabetes and obesity through augmentation of postprandial GLP-1 secretion by CAR8 inhibition.


Subject(s)
Biomarkers, Tumor/metabolism , Corn Oil/pharmacology , Enteroendocrine Cells/drug effects , Fatty Acids/pharmacology , Glucagon-Like Peptide 1/metabolism , Nerve Tissue Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Calcium Signaling , Cell Line , Enteroendocrine Cells/enzymology , Glucagon/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Secretory Pathway , Type C Phospholipases/metabolism
2.
Cell Calcium ; 93: 102327, 2021 01.
Article in English | MEDLINE | ID: mdl-33316585

ABSTRACT

Inositol polyphosphate multikinase (IPMK) is a conserved protein that initiates the production of inositol phosphate intracellular messengers and is critical for regulating a variety of cellular processes. Here, we report that the C. elegans IPMK-1, which is homologous to the mammalian inositol polyphosphate multikinase, plays a crucial role in regulating rhythmic behavior and development. The deletion mutant ipmk-1(tm2687) displays a long defecation cycle period and retarded postembryonic growth. The expression of functional ipmk-1::GFP was detected in the pharyngeal muscles, amphid sheath cells, the intestine, excretory (canal) cells, proximal gonad, and spermatheca. The expression of IPMK-1 in the intestine was sufficient for the wild-type phenotype. The IP3-kinase activity of IPMK-1 is required for defecation rhythms and postembryonic development. The defective phenotypes of ipmk-1(tm2687) could be rescued by a loss-of-function mutation in type I inositol 5-phosphatase homolog (IPP-5) and improved by a supplemental Ca2+ in the medium. Our work demonstrates that IPMK-1 and the signaling molecule inositol triphosphate (IP3) pathway modulate rhythmic behaviors and development by dynamically regulating the concentration of intracellular Ca2+ in C. elegans. Advances in understanding the molecular regulation of Ca2+ homeostasis and regulation of organism development may lead to therapeutic strategies that modulate Ca2+ signaling to enhance function and counteract disease processes. Unraveling the physiological role of IPMK and the underlying functional mechanism in C. elegans would contribute to understanding the role of IPMK in other species, especially in mammals, and benefit further research on the involvement of IPMK in disease.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Caenorhabditis elegans/enzymology , Calcium Signaling , Embryonic Development , Inositol 1,4,5-Trisphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Defecation , Gene Deletion , Intracellular Space/metabolism , Mutation/genetics , Organ Specificity , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/chemistry
3.
Genome ; 63(2): 61-90, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31557446

ABSTRACT

Nucleobindin-1 is an EF-hand calcium-binding protein with a distinctive profile, predominantly localized to the Golgi in insect and wide-ranging vertebrate cell types, alike. Its putative involvements in intracellular calcium (Ca2+) homeostasis have never been phenotypically characterized in any model organism. We have analyzed an adult-viable mutant that completely disrupts the G protein α-subunit binding and activating (GBA) motif of Drosophila Nucleobindin-1 (dmNUCB1). Such disruption does not manifest any obvious fitness-related, morphological/developmental, or behavioral abnormalities. A single copy of this mutation or the knockdown of dmnucb1 in restricted sets of cells variously rescues pleiotropic mutant phenotypes arising from impaired inositol 1,4,5-trisphosphate receptor (IP3R) activity (in turn depleting cytoplasmic Ca2+ levels across diverse tissue types). Additionally, altered dmNUCB1 expression or function considerably reverses lifespan and mobility improvements effected by IP3R mutants, in a Drosophila model of amyotrophic lateral sclerosis. Homology modeling-based analyses further predict a high degree of conformational conservation in Drosophila, of biochemically validated structural determinants in the GBA motif that specify in vertebrates, the unconventional Ca2+-regulated interaction of NUCB1 with Gαi subunits. The broad implications of our findings are hypothetically discussed, regarding potential roles for NUCB1 in GBA-mediated, Golgi-associated Ca2+ signaling, in health and disease.


Subject(s)
Calcium-Binding Proteins/physiology , Calcium/metabolism , Drosophila Proteins/physiology , Inositol 1,4,5-Trisphosphate Receptors/genetics , Nucleobindins/physiology , Alleles , Amino Acid Motifs , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Genes, Lethal , Genetic Pleiotropy , Golgi Apparatus/metabolism , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Mutation , Nucleobindins/chemistry , Nucleobindins/genetics , Nucleobindins/metabolism , Protein Domains , Structural Homology, Protein
4.
Int J Biol Macromol ; 124: 1321-1327, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30244130

ABSTRACT

IP3 is a ubiquitous second messenger in eukaryotic cells that triggers Ca2+-release from intracellular stores. IP3 binds to intracellular IP3-receptor (IP3R) and induces conformational change within the ligand-binding domain which regulates Ca2+-release; hence, both IP3 and IP3R are key components of the signal transduction mechanism. Here we present cDNA cloning of IP3-binding core (IBC) domain encoding only residues 224-604 of human IP3R type 2 that binds to IP3 with high affinity. RNA extraction, RT-PCR, PCR and cloning were carried out, and then the cloned DNA was checked by sequencing. Thereafter, expression vector pET-28a harboring the correct gene was transformed into different E. coli (DE3) strains and investigated its protein expression under various conditions. Finally, the IBC expression was induced at 20 °C for 20 h into BL21 strain at LB medium with 4 mM lactose and 0.5 mM IPTG, and then confirmed by western blotting. After protein purification, structural study was recorded in absence and presence of its ligand. Far-CD and intrinsic fluorescence spectra analysis of the purified protein with and without IP3 ligand showed change in secondary and tertiary IBC structure. Moreover, bioinformatics study demonstrated that the ligand binding site residues R269, K508 and R511 are conserved.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/isolation & purification , Inositol 1,4,5-Trisphosphate/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Computational Biology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Ligands , Mice , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
5.
J Biol Chem ; 293(34): 13112-13124, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29970616

ABSTRACT

Fine-tuning of the activity of inositol 1,4,5-trisphosphate receptors (IP3R) by a diverse array of regulatory inputs results in intracellular Ca2+ signals with distinct characteristics. These events allow the activation of specific downstream effectors. We reported previously that region-specific proteolysis represents a novel regulatory event for type 1 IP3R (R1). Specifically, caspase-fragmented R1 display a marked increase in single-channel open probability. More importantly, the distinct characteristics of the Ca2+ signals elicited via fragmented R1 can activate alternate downstream effectors. In this report, we expand these studies to investigate whether all IP3R subtypes are regulated by proteolysis. We now show that type 2 and type 3 IP3R (R2 and R3, respectively) are proteolytically cleaved in rodent models of acute pancreatitis. Surprisingly, fragmented IP3R retained tetrameric architecture, remained embedded in endoplasmic reticulum membranes and were not functionally disabled. Proteolysis was associated with a marked attenuation of the frequency of Ca2+ signals in pancreatic lobules. Consistent with these data, expression of DNAs encoding complementary R2 and R3 peptides mimicking fragmented receptors at particular sites, resulted in a significant decrease in the frequency of agonist-stimulated Ca2+ oscillations. Further, proteolysis of R2 resulted in a marked decrease in single-channel open probability. Taken together, proteolytic fragmentation modulates R2 and R3 activity in a region-specific manner, and this event may contribute to the altered Ca2+ signals in pancreatic acinar cells during acute pancreatitis.


Subject(s)
Calcium Signaling , Disease Models, Animal , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Pancreatitis/physiopathology , Acute Disease , Animals , Inositol 1,4,5-Trisphosphate Receptors/genetics , Ion Channel Gating , Male , Mice , Mice, Inbred C57BL , Proteolysis , Rats , Rats, Wistar
6.
Bioelectromagnetics ; 38(6): 436-446, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28570746

ABSTRACT

This research investigated the influence of extremely low frequency magnetic fields (ELF-MF; 50 Hz, 8 mT, 4 h per day, for 28 days) on calcium ion signaling and the double messenger system in the hippocampus of mice. Messengers that were studied included: G-protein, Ins(1,4,5)P3 (IP3 ), diacylglycerol (DAG), cAMP-dependent protein kinase (PKA), and Ca2+ -dependent protein kinase C (PKC). The results showed that ELF-MF caused an increase in the levels of Gi protein, IP3, DAG, PKA and PKC beta, calcium and calmodulin-dependent protein phosphatase calcineuring (PP2B), and intracellular Ca2+ content, and a decrease in calcium/calmodulin-dependent protein kinase II (CaMK II) and PKC alpha. In addition, ELF-MF exposure decreased the level of brain-derived neurotrophic factor (BDNF), which played a key role in hippocampal neuronal cell death. However, oral administration of procyanidins from lotus seedpod (LSPCs) (especially 90 mg kg-1 ) significantly recovered these changes, and nearly reached normal levels. All these showed that LSPCs may mediate calcium signal and double messenger system through Ca2+ /CaMK II/CREB/BDNF and DG/PKC/MAPK signaling pathways to reverse the alteration caused by ELF-MF. Bioelectromagnetics. 38:436-446, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Biflavonoids/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/radiation effects , Catechin/pharmacology , Hippocampus/cytology , Magnetic Fields/adverse effects , Magnoliopsida/chemistry , Proanthocyanidins/pharmacology , Seeds/chemistry , Animals , Biflavonoids/isolation & purification , Brain-Derived Neurotrophic Factor/metabolism , Catechin/isolation & purification , Diglycerides/metabolism , GTP-Binding Proteins/metabolism , Hippocampus/drug effects , Hippocampus/radiation effects , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice , Mice, Inbred ICR , Proanthocyanidins/isolation & purification , Protein Kinases/metabolism
7.
J Biol Chem ; 292(28): 11714-11726, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28526746

ABSTRACT

The inositol 1,4,5 trisphosphate receptor (IP3R) is an intracellular Ca2+ release channel expressed predominately on the membranes of the endoplasmic reticulum. IP3R1 can be cleaved by caspase or calpain into at least two receptor fragments. However, the functional consequences of receptor fragmentation are poorly understood. Our previous work has demonstrated that IP3R1 channels, formed following either enzymatic fragmentation or expression of the corresponding complementary polypeptide chains, retain tetrameric architecture and are still activated by IP3 binding despite the loss of peptide continuity. In this study, we demonstrate that region-specific receptor fragmentation modifies channel regulation. Specifically, the agonist-evoked temporal Ca2+ release profile and protein kinase A modulation of Ca2+ release are markedly altered. Moreover, we also demonstrate that activation of fragmented IP3R1 can result in a distinct functional outcome. Our work suggests that proteolysis of IP3R1 may represent a novel form of modulation of IP3R1 channel function and increases the repertoire of Ca2+ signals achievable through this channel.


Subject(s)
Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Protein Processing, Post-Translational , Amino Acid Substitution , Animals , Cell Line , Chickens , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Knockout Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/genetics , Kinetics , Mutation , Patch-Clamp Techniques , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Proteolysis , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Up-Regulation
8.
PLoS One ; 12(4): e0175778, 2017.
Article in English | MEDLINE | ID: mdl-28448505

ABSTRACT

In recent years, by extensive achievements in understanding the mechanisms and the pathways affected by cancer, the focus of cancer research is shifting from developing new chemotherapy methods to using natural compounds with therapeutic properties to reduce the adverse effects of synthetic drugs on human health. We used fruit extracts from previously generated human type I InsP 5-ptase gene expressing transgenic tomato plants for assessment of the anti-cancer activity of established genetically modified tomato lines. Cellular assays (MTT, Fluorescent microscopy, Flow Cytometry analysis) were used to confirm that InsP 5-ptase fruit extract was more effective for reducing the proliferation of breast cancer cells compared to wild-type tomato fruit extract. Metabolome analysis of InsP 5-ptase expressing tomato fruits performed by LC-MS identified tomato metabolites that may play a key role in the increased anti-cancer activity observed for the transgenic fruits. Total transcriptome analysis of cancer cells (MCF-7 line) exposed to an extract of transgenic fruits revealed a number of differently regulated genes in the cells treated with transgenic extract compared to untreated cells or cells treated with wild-type tomato extract. Together, this data demonstrate the potential role of the plant derived metabolites in suppressing cell viability of cancer cells and further prove the potential application of plant genetic engineering in the cancer research and drug discovery.


Subject(s)
Cell Proliferation/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , Plant Extracts/chemistry , Plant Proteins/metabolism , Polyphenols/toxicity , Solanum lycopersicum/chemistry , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Flavonoids/analysis , Flavonoids/metabolism , Flow Cytometry , Fruit/chemistry , Fruit/metabolism , Humans , Solanum lycopersicum/metabolism , MCF-7 Cells , Mass Spectrometry , Metabolome , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Plants, Genetically Modified/chemistry , Plants, Genetically Modified/metabolism , Polyphenols/chemistry , Polyphenols/isolation & purification , Transcriptome/drug effects
9.
Lasers Med Sci ; 32(1): 169-180, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27864646

ABSTRACT

Low-level laser (LLL) irradiation has been reported to promote neuronal differentiation, but the mechanism remains unclear. Brain-derived neurotrophic factor (BDNF) has been confirmed to be one of the most important neurotrophic factors because it is critical for the differentiation and survival of neurons during development. Thus, this study aimed to investigate the effects of LLL irradiation on Bdnf messenger RNA (mRNA) transcription and the molecular pathway involved in LLL-induced Bdnf mRNA transcription in cultured dorsal root ganglion neurons (DRGNs) using Ca2+ imaging, pharmacological detections, RNA interference, immunocytochemistry assay, Western blot, and qPCR analysis. We show here that LLL induced increases in the [Ca2+] i level, Bdnf mRNA transcription, cAMP-response element-binding protein (CREB) phosphorylation, and extracellular signal-regulated kinase (ERK) phosphorylation, mediated by Ca2+ release via inositol triphosphate receptor (IP3R)-sensitive calcium (Ca2+) stores. Blockade of Ca2+ increase suppressed Bdnf mRNA transcription, CREB phosphorylation, and ERK phosphorylation. Downregulation of phosphorylated (p)-CREB reduced Bdnf mRNA transcription triggered by LLL. Furthermore, blockade of ERK using PD98059 inhibitor reduced p-CREB and Bdnf mRNA transcription induced by LLL. Taken together, these findings establish the Ca2+-ERK-CREB cascade as a potential signaling pathway involved in LLL-induced Bdnf mRNA transcription. To our knowledge, this is the first report of the mechanisms of Ca2+-dependent Bdnf mRNA transcription triggered by LLL. These findings may help further explore the complex molecular signaling networks in LLL-triggered nerve regeneration in vivo and may also provide experimental evidence for the development of LLL for clinical applications.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Low-Level Light Therapy , Signal Transduction/radiation effects , Transcription, Genetic , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Survival/radiation effects , Cells, Cultured , Enzyme Activation/radiation effects , Inositol 1,4,5-Trisphosphate/metabolism , Models, Biological , Neurogenesis/radiation effects , Neurons/cytology , Neurons/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Transcription, Genetic/radiation effects
10.
Int J Food Sci Nutr ; 67(4): 454-60, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27019314

ABSTRACT

Commercial preparations of 6-phytase A alone and in combination with phytase B were used in rye breadmaking. Determination of bioavailability of myo-inositol phosphates from bread was performed by an in vitro digestion method followed by the measurement of an uptake by Caco-2 cells in culture. In bread supplemented with a combination of 6-phytase A and phytase B, a significant reduction in phytate content was observed from 3.62 µmol/g in the control to 0.7 µmol/g. Bioavailability of phytate estimated by an in vitro method simulating digestion in the human alimentary tract was 9% in the bread supplemented with phytase B, 7% (6-phytase A) and 50% in the control bread. In cell culture, the bioaccessibilities of inositol triphosphates from bread baked with the addition of 6-phytase A was higher by 36% as compared to the samples baked with phytase B and by 32% in breads baked with combination of both phytases.


Subject(s)
6-Phytase/metabolism , Bread/analysis , Dietary Fiber/metabolism , Food Additives/metabolism , Inositol Phosphates/metabolism , Intestinal Absorption , Secale/chemistry , Anion Exchange Resins , Caco-2 Cells , Chromatography, High Pressure Liquid , Dietary Fiber/analysis , Digestion , Flour/analysis , Humans , Inositol 1,4,5-Trisphosphate/analysis , Inositol 1,4,5-Trisphosphate/metabolism , Inositol Phosphates/analysis , Isoenzymes , Models, Biological , Nutritive Value , Phosphorylation , Phytic Acid/analysis , Phytic Acid/metabolism , Poland
11.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L747-58, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26773068

ABSTRACT

Enhanced contractility of airway smooth muscle (ASM) is a major pathophysiological characteristic of asthma. Expanding the therapeutic armamentarium beyond ß-agonists that target ASM hypercontractility would substantially improve treatment options. Recent studies have identified naturally occurring phytochemicals as candidates for acute ASM relaxation. Several flavonoids were evaluated for their ability to acutely relax human and murine ASM ex vivo and murine airways in vivo and were evaluated for their ability to inhibit procontractile signaling pathways in human ASM (hASM) cells. Two members of the flavonol subfamily, galangin and fisetin, significantly relaxed acetylcholine-precontracted murine tracheal rings ex vivo (n = 4 and n = 5, respectively, P < 0.001). Galangin and fisetin also relaxed acetylcholine-precontracted hASM strips ex vivo (n = 6-8, P < 0.001). Functional respiratory in vivo murine studies demonstrated that inhaled galangin attenuated the increase in lung resistance induced by inhaled methacholine (n = 6, P < 0.01). Both flavonols, galangin and fisetin, significantly inhibited purified phosphodiesterase-4 (PDE4) (n = 7, P < 0.05; n = 7, P < 0.05, respectively), and PLCß enzymes (n = 6, P < 0.001 and n = 6, P < 0.001, respectively) attenuated procontractile Gq agonists' increase in intracellular calcium (n = 11, P < 0.001), acetylcholine-induced increases in inositol phosphates, and CPI-17 phosphorylation (n = 9, P < 0.01) in hASM cells. The prorelaxant effect retained in these structurally similar flavonols provides a novel pharmacological method for dual inhibition of PLCß and PDE4 and therefore may serve as a potential treatment option for acute ASM constriction.


Subject(s)
Flavonoids/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Phospholipase C beta/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/physiopathology , Asthma/drug therapy , Bronchoconstriction/drug effects , Calcium Signaling , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Drug Evaluation, Preclinical , Flavonoids/chemistry , Flavonols , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice , Muscle Contraction , Muscle, Smooth/physiology , Muscle, Smooth/physiopathology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Phospholipase C beta/physiology
12.
J Physiol ; 594(11): 2867-76, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26486785

ABSTRACT

Inositol 1,4,5-trisphosphate receptors (IP3 Rs) are a family of ubiquitously expressed intracellular Ca(2+) release channels. Regulation of channel activity by Ca(2+) , nucleotides, phosphorylation, protein binding partners and other cellular factors is thought to play a major role in defining the specific spatiotemporal characteristics of intracellular Ca(2+) signals. These properties are, in turn, believed pivotal for the selective and specific physiological activation of Ca(2+) -dependent effectors. IP3 Rs are also substrates for the intracellular cysteine proteases, calpain and caspase. Cleavage of the IP3 R has been proposed to play a role in apoptotic cell death by uncoupling regions important for IP3 binding from the channel domain, leaving an unregulated leaky Ca(2+) pore. Contrary to this hypothesis, we demonstrate following proteolysis that N- and C-termini of IP3 R1 remain associated, presumably through non-covalent interactions. Further, we show that complementary fragments of IP3 R1 assemble into tetrameric structures and retain their ability to be regulated robustly by IP3 . While peptide continuity is clearly not necessary for IP3 -gating of the channel, we propose that cleavage of the IP3 R peptide chain may alter other important regulatory events to modulate channel activity. In this scenario, stimulation of the cleaved IP3 R may support distinct spatiotemporal Ca(2+) signals and activation of specific effectors. Notably, in many adaptive physiological events, the non-apoptotic activities of caspase and calpain are demonstrated to be important, but the substrates of the proteases are poorly defined. We speculate that proteolytic fragmentation may represent a novel form of IP3 R regulation, which plays a role in varied adaptive physiological processes.


Subject(s)
Calcium Channels/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Humans , Protein Binding/physiology , Proteolysis
13.
Peptides ; 75: 8-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26524625

ABSTRACT

Bombesin receptor subtype-3 (BRS-3) is an orphan G-protein coupled receptor which is classified in the bombesin receptor (BnR) family with which it shares high homology. It is present widely in the central nervous system and peripheral tissues and primarily receptor-knockout studies suggest it is involved in metabolic-glucose-insulin homeostasis, feeding and other CNS behaviors, gastrointestinal motility and cancer growth. However, the role of BRS-3 physiologically or in pathologic disorders has been not well defined because the natural ligand is unknown. Until recently, no selective agonists/antagonists were available; however, recently synthetic high-affinity agonists, chiral-diazepines nonpeptide-analogs (3F, 9D, 9F, 9G) with low CNS penetrance, were described, but are not well-categorized pharmacologically or in different labarotory species. The present study characterizes the affinities, potencies, selectivities of the chiral-diazepine BRS-3 agonists in human and rodents (mice,rat). In human BRS-3 receptors, the relative affinities of the chiral-diazepines was 9G>9D>9F>3F; each was selective for BRS-3. For stimulating PLC activity, in h-BRS-3 each of the four chiral diazepine analogs was fully efficacious and their relative potencies were: 9G (EC50: 9 nM)>9D (EC50: 9.4 nM)>9F (EC50: 39 nM)>3F (EC50: 48 nM). None of the four chiral diazepine analogs activated r,m,h-GRPR/NMBR. The nonpeptide agonists showed marked differences from each other and a peptide agonist in receptor-coupling-stiochiometry and in affinities/potencies in different species. These results demonstrate that chiral diazepine analogs (9G, 9D, 9F, 3F) have high/affinity/potency for the BRS-3 receptor in human and rodent cells, but different coupling-relationships and species differences from a peptide agonist.


Subject(s)
Azepines/pharmacology , Receptors, Bombesin/agonists , Animals , BALB 3T3 Cells , Drug Evaluation, Preclinical , Gastrin-Releasing Peptide/pharmacology , Humans , Inhibitory Concentration 50 , Inositol 1,4,5-Trisphosphate/metabolism , Mice , Neurokinin B/analogs & derivatives , Neurokinin B/pharmacology , Rats , Second Messenger Systems , Species Specificity
14.
J Ethnopharmacol ; 175: 422-31, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26429073

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Alstonia scholaris has a long history of use in the Ayurveda traditional treatment of various ailments including hypertension. We have reported the blood pressure lowering activity of the extract of A. scholaris. The following research aim to delineate the pharmacological mechanism involve in the antihypertensive action. MATERIALS AND METHOD: Vasorelaxant effect of the n-butanol fraction of A. scholaris (NBF-ASME) was evaluated on rat aorta pre-contracted with phenyelphrine (PE, 1 µM). Aortic rings preparation were pre-incubated with various antagonists like 1H-[1,2,4] oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ 10 µM), methylene blue (MB 10 µM), Nω-nitro-L-arginine methyl ester hydrochloride (l-NAME 10 µM), atropine (10 µM), indomethacin (1 µM), ML-9 and various K(+) channel blockers such as glibenclamide (10 µM) and tetraethyl ammonium (TEA 10 µM) for mechanism study. RESULT: The results showed that pre-incubation of aortic rings with the extract (0.5, 1 and 2mg/mL) significantly inhibit the contractile response of the rings to phenylephrine-induced contraction (p<0.05-0.001). Removal of endothelium, incubation with L-NAME, indomethacin, atropine and propranolol did not significantly affect the relaxation effect of NBF-ASME. Furthermore, the K(+) channel blockers, TEA and glibenclamide showed no inhibitory effect. However, aortic rings pretreated with ODQ and ML-9 showed a significant suppression of the relaxation curve of NBF-ASME (p<0.01-0.001). In Ca(2+)-free solution, NBF-ASME inhibits the release of intracellular Ca(2+) from the sarcoplasmic reticulum. NBF-ASME also inhibits calcium chloride (CaCl2)-induced contraction in endothelium-denuded aortic rings. CONCLUSION: The results from this study suggests that A. scholaris exerts vasodilation via calcium channels blockade, direct activation of soluble guanylate cyclase and possibly by also inhibiting the formation of inositol 1, 4, 5-triphosphate.


Subject(s)
Alstonia , Antihypertensive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Plant Extracts/pharmacology , Vasodilator Agents/pharmacology , 1-Butanol/chemistry , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Muscarinic Antagonists/pharmacology , Nitric Oxide/metabolism , Plant Bark , Potassium Channel Blockers/pharmacology , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Solvents/chemistry , Vasoconstriction/drug effects
15.
Am J Pathol ; 185(4): 1061-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25794706

ABSTRACT

During cholestatic liver disease, there is dysregulation in the balance between biliary growth and loss in bile duct-ligated (BDL) rats modulated by neuroendocrine peptides via autocrine/paracrine pathways. Gonadotropin-releasing hormone (GnRH) is a trophic peptide hormone that modulates reproductive function and proliferation in many cell types. We evaluated the autocrine role of GnRH in the regulation of cholangiocyte proliferation. The expression of GnRH receptors was assessed in a normal mouse cholangiocyte cell line (NMC), sham, and BDL rats. The effect of GnRH administration was evaluated in normal rats and in NMC. GnRH-induced biliary proliferation was evaluated by changes in intrahepatic bile duct mass and the expression of proliferation and function markers. The expression and secretion of GnRH in NMC and isolated cholangiocytes was assessed. GnRH receptor subtypes GnRHR1 and GnRHR2 were expressed in cholangiocytes. Treatment with GnRH increased intrahepatic bile duct mass as well as proliferation and function markers in cholangiocytes. Transient knockdown and pharmacologic inhibition of GnRHR1 in NMC decreased proliferation. BDL cholangiocytes had increased expression of GnRH compared with normal rats, accompanied by increased GnRH secretion. In vivo and in vitro knockdown of GnRH decreased intrahepatic bile duct mass/cholangiocyte proliferation and fibrosis. GnRH secreted by cholangiocytes promotes biliary proliferation via an autocrine pathway. Disruption of GnRH/GnRHR signaling may be important for the management of cholestatic liver diseases.


Subject(s)
Autocrine Communication , Bile Ducts, Intrahepatic/cytology , Gonadotropin-Releasing Hormone/metabolism , Paracrine Communication , Animals , Bile Ducts, Intrahepatic/drug effects , Cell Line , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Fluorescent Antibody Technique , Gene Silencing/drug effects , Hypothalamus/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Liver Cirrhosis/pathology , Male , Mice , Morpholinos/administration & dosage , Morpholinos/pharmacology , Paracrine Communication/drug effects , Rats, Inbred F344 , Receptors, LHRH/metabolism
16.
Mol Cell Endocrinol ; 406: 1-9, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25697345

ABSTRACT

Intracellular Ca(2+) signaling is important for stem cell differentiation and there is evidence it may coordinate the process. Arginine vasopressin (AVP) is a neuropeptide hormone secreted mostly from the posterior pituitary gland and increases Ca(2+) signals mainly via V1 receptors. However, the role of AVP in adipogenesis of human adipose-derived stem cells (hASCs) is unknown. In this study, we identified the V1a receptor gene in hASCs and demonstrated that AVP stimulation increased intracellular Ca(2+) concentration during adipogenesis. This effect was mediated via V1a receptors, Gq-proteins and the PLC-IP3 pathway. These Ca(2+) signals were due to endoplasmic reticulum release and influx from the extracellular space. Furthermore, AVP supplementation to the adipogenic medium decreased the number of adipocytes and adipocyte marker genes during differentiation. The effect of AVP on adipocyte formation was reversed by the V1a receptor blocker V2255. These findings suggested that AVP may function to inhibit adipocyte differentiation.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/cytology , Arginine Vasopressin/pharmacology , Stem Cells/cytology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Aged , Antidiuretic Hormone Receptor Antagonists/pharmacology , Arginine Vasopressin/analogs & derivatives , Calcium/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , Female , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Intracellular Space/metabolism , Male , Middle Aged , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Type C Phospholipases/metabolism
17.
Chin J Integr Med ; 21(1): 29-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24817315

ABSTRACT

OBJECTIVE: To investigate the effect of Chaiqin Chengqi Decoction (,CQCQD) on cholecystokinin receptor 1 (CCKR1)-mediated signal transduction of pancreatic acinar cell in rats with acute necrotic pancreatitis (ANP). METHODS: Twenty-seven Sprague-Dawley rats were randomized into three groups: the control group, the ANP group, and the CQCQD group (9 in each group). ANP rats were induced by two intraperitoneal injections of 8% L-arginine (pH=7.0, 4.4 g/kg) over a 2-h period. Rats were treated with 1.5 mL/100 g body weight of CQCQD (CQCQD group) or physiological saline (control and ANP groups) at 2 h interval. And 6 h after induction, pancreatic tissues were collected for histopathological examination. Pancreatic acinar cells were isolated for determination of CCKR1 mRNA and protein expression, phospholipase C (PLC) and inositol-1,4,5-triphosphate (IP3), and determination of fluorescence intensity (FI) as a measure of intracellular calcium ion concentration [Ca(2+)]i. RESULTS: The pancreatic histopathological score (6.2 ± 1.1) and the levels of PLC (1,187.2 ± 228.2 µg/mL) and IP3 (872.2 ± 88.4 µg/mL) of acinar cells in the ANP group were higher than those in the control (2.8 ± 0.4, 682.5 ± 121.8 µg/mL, 518.4 ± 115.8 µg/mL) and the CQCQD (3.8 ± 0.8, 905.3 ± 78.5 µg/mL, 611.0 ± 42.5 µg/mL) groups (P<0.05). [Ca(2+)]i FI for the ANP group (34.8±27.0) was higher than that in the control (5.1 ± 2.2) and CQCQD (12.6 ± 2.5) groups (P<0.05). The expression of pancreatic acinar cell CCKR1 mRNA in the ANP group was up-regulated (expression ratio=1.761; P=0.024) compared with the control group. The expression of pancreatic acinar cell CCKR1 mRNA in the CQCQD group was down-regulated (expression ratio=0.311; P=0.035) compared with the ANP group. The ratio of gray values of the CCKR1 and ß-actin in the ANP group (1.43 ± 0.17) was higher than those in the control (0.70 ± 0.15) and CQCQD (0.79 ± 0.11) groups (P<0.05). CONCLUSIONS: Pancreatic acinar cell calcium overload of ANP induced by L-arginine was related to the up-regulated expressions of pancreatic acinar cell CCKR1 mRNA and protein. CQCQD can down-regulate expressions of pancreatic acinar cell CCKR1 mRNA and protein to reduce the PLC and IP3 of pancreatic acinar cells, relieving the calcium overload and reducing the pathological changes in rats with ANP.


Subject(s)
Acinar Cells/metabolism , Drugs, Chinese Herbal/therapeutic use , Pancreas/pathology , Pancreatitis, Acute Necrotizing/drug therapy , Pancreatitis, Acute Necrotizing/pathology , Receptors, Cholecystokinin/metabolism , Signal Transduction , Acinar Cells/drug effects , Animals , Blotting, Western , Calcium/metabolism , Drugs, Chinese Herbal/pharmacology , Fluorescence , Gene Expression Regulation/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Cholecystokinin/genetics , Signal Transduction/drug effects , Type C Phospholipases/metabolism
18.
Structure ; 22(10): 1446-57, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295397

ABSTRACT

We investigated the mechanisms of activation and degradation of the E3 ubiquitin ligase Nedd4L combining the available biochemical information with complementary biophysical techniques. Using nuclear magnetic resonance spectroscopy, we identified that the C2 domain binds Ca(2+) and inositol 1,4,5-trisphosphate (IP3) using the same interface that is used to interact with the HECT domain. Thus, we propose that the transition from the closed to the active form is regulated by a competition of IP3 and Ca(2+) with the HECT domain for binding to the C2 domain. We performed relaxation experiments and molecular dynamic simulations to determine the flexibility of the HECT structure and observed that its conserved PY motif can become solvent-exposed when the unfolding process is initiated. The structure of the WW3 domain bound to the HECT-PY site reveals the details of this interaction, suggesting a possible auto-ubquitination mechanism using two molecules, a partially unfolded one and a fully functional Nedd4L counterpart.


Subject(s)
Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Binding, Competitive , Calcium/metabolism , Enzyme Activation , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Protein Conformation , Protein Structure, Tertiary , Signal Transduction , Ubiquitination
19.
Am J Physiol Cell Physiol ; 306(8): C736-44, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24401846

ABSTRACT

The existence of a local renin-angiotensin system (RAS) in neurons was first postulated 40 years ago. Further studies indicated intraneuronal generation of ANG II. However, the function and signaling mechanisms of intraneuronal ANG II remained elusive. Since ANG II type 1 receptor (AT1R) is the major type of receptor mediating the effects of ANG II, we used intracellular microinjection and concurrent Ca(2+) and voltage imaging to examine the functionality of intracellular AT1R in neurons. We show that intracellular administration of ANG II produces a dose-dependent elevation of cytosolic Ca(2+) concentration ([Ca(2+)]i) in hypothalamic neurons that is sensitive to AT1R antagonism. Endolysosomal, but not Golgi apparatus, disruption prevents the effect of microinjected ANG II on [Ca(2+)]i. Additionally, the ANG II-induced Ca(2+) response is dependent on microautophagy and sensitive to inhibition of PLC or antagonism of inositol 1,4,5-trisphosphate receptors. Furthermore, intracellular application of ANG II produces AT1R-mediated depolarization of hypothalamic neurons, which is dependent on [Ca(2+)]i increase and on cation influx via transient receptor potential canonical channels. In summary, we provide evidence that intracellular ANG II activates endolysosomal AT1Rs in hypothalamic neurons. Our results point to the functionality of a novel intraneuronal angiotensinergic pathway, extending the current understanding of intracrine ANG II signaling.


Subject(s)
Angiotensin II/metabolism , Neurons/physiology , Signal Transduction/physiology , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Gene Expression Regulation , Humans , Hypothalamus/cytology , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Microinjections , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism
20.
PLoS One ; 9(1): e85937, 2014.
Article in English | MEDLINE | ID: mdl-24465800

ABSTRACT

G protein-coupled receptors (GPCRs) show some level of basal activity even in the absence of an agonist, a phenomenon referred to as constitutive activity. Such constitutive activity in GPCRs is known to have important pathophysiological roles in human disease. The thromboxane A2 receptor (TP) is a GPCR that promotes thrombosis in response to binding of the prostanoid, thromboxane A2. TP dysfunction is widely implicated in pathophysiological conditions such as bleeding disorders, hypertension and cardiovascular disease. Recently, we reported the characterization of a few constitutively active mutants (CAMs) in TP, including a genetic variant A160T. Using these CAMs as reporters, we now test the inverse agonist properties of known antagonists of TP, SQ 29,548, Ramatroban, L-670596 and Diclofenac, in HEK293T cells. Interestingly, SQ 29,548 reduced the basal activity of both, WT-TP and the CAMs while Ramatroban was able to reduce the basal activity of only the CAMs. Diclofenac and L-670596 showed no statistically significant reduction in basal activity of WT-TP or CAMs. To investigate the role of these compounds on human platelet function, we tested their effects on human megakaryocyte based system for platelet activation. Both SQ 29,548 and Ramatroban reduced the platelet hyperactivity of the A160T genetic variant. Taken together, our results suggest that SQ 29,548 and Ramatroban are inverse agonists for TP, whereas, L-670596 and Diclofenac are neutral antagonists. Our findings have important therapeutic applications in the treatment of TP mediated pathophysiological conditions.


Subject(s)
Carbazoles/pharmacology , Hydrazines/pharmacology , Receptors, Thromboxane A2, Prostaglandin H2/agonists , Sulfonamides/pharmacology , Amino Acid Substitution , Blood Platelets/drug effects , Blood Platelets/metabolism , Bridged Bicyclo Compounds, Heterocyclic , Calcium Signaling/drug effects , Drug Evaluation, Preclinical , Fatty Acids, Unsaturated , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Mutagenesis, Site-Directed , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL