Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Physiol Sci ; 71(1): 2, 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33461483

ABSTRACT

BACKGROUND: We previously established that the non-neuronal cardiac cholinergic system (NNCCS) is equipped with cardiomyocytes synthesizes acetylcholine (ACh), which is an indispensable endogenous system, sustaining cardiac homeostasis and regulating an inflammatory status, by transgenic mice overexpressing choline acetyltransferase (ChAT) gene in the heart. However, whole body biological significances of NNCCS remain to be fully elucidated. METHODS AND RESULTS: To consolidate the features, we developed heart-specific ChAT knockdown (ChATKD) mice using 3 ChAT-specific siRNAs. The mice developed cardiac dysfunction. Factors causing it included the downregulation of cardiac glucose metabolism along with decreased signal transduction of Akt/HIF-1alpha/GLUT4, leading to poor glucose utilization, impairment of glycolytic metabolites entering the tricarboxylic (TCA) cycle, the upregulation of reactive oxygen species (ROS) production with an attenuated scavenging potency, and the downregulated nitric oxide (NO) production via NOS1. ChATKD mice revealed a decreased vagus nerve activity, accelerated aggression, more accentuated blood basal corticosterone levels with depression-like phenotypes, several features of which were accompanied by cardiac dysfunction. CONCLUSION: The NNCCS plays a crucial role in cardiac homeostasis by regulating the glucose metabolism, ROS synthesis, NO levels, and the cardiac vagus nerve activity. Thus, the NNCCS is suggested a fundamentally crucial system of the heart.


Subject(s)
Acetylcholine/metabolism , Choline O-Acetyltransferase/metabolism , Myocardium/metabolism , Animals , Blood Pressure , Choline O-Acetyltransferase/genetics , Down-Regulation , Gene Expression Regulation, Enzymologic , Histones/genetics , Histones/metabolism , Homeostasis , Malondialdehyde , Mice , Mice, Transgenic , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/genetics , Tyrosine/metabolism
2.
Int Immunopharmacol ; 84: 106459, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32325404

ABSTRACT

We have previously reported the development of a novel chemical compound, S-Nitroso-N-Pivaloyl-D-Penicillamine (SNPiP), for the upregulation of the non-neuronal cardiac cholinergic system (NNCCS), a cardiac acetylcholine (ACh) synthesis system, which is different from the vagus nerve releasing of ACh as a neurotransmitter. However, it remains unclear how SNPiP could influence cardiac function positively, and whether SNPiP could improve cardiac function under various pathological conditions. SNPiP-injected control mice demonstrated a gradual upregulation in diastolic function without changes in heart rate. In contrast to some parameters in cardiac function that were influenced by SNPiP 24 h or 48 h after a single intraperitoneal (IP) injection, 72 h later, end-systolic pressure, cardiac output, end-diastolic volume, stroke volume, and ejection fraction increased. IP SNPiP injection also improved impaired cardiac function, which is a characteristic feature of the db/db heart, in a delayed fashion, including diastolic and systolic function, following either several consecutive injections or a single injection. SNPiP, a novel NNCCS activator, could be applied as a therapeutic agent for the upregulation of NNCCS and as a unique tool for modulating cardiac function via improvement in diastolic function.


Subject(s)
Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Heart/drug effects , Nitric Oxide Donors/pharmacology , Non-Neuronal Cholinergic System/physiology , Penicillamine/pharmacology , Ventricular Function, Left/drug effects , Animals , Blood Pressure/drug effects , Injections, Intraperitoneal , Injections, Intravenous , Mice , Mice, Inbred Strains , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/therapeutic use , Penicillamine/administration & dosage , Penicillamine/analogs & derivatives , Penicillamine/therapeutic use
3.
Brain Behav Immun ; 81: 122-137, 2019 10.
Article in English | MEDLINE | ID: mdl-31176726

ABSTRACT

We previously reported that the heart-specific choline acetyltransferase (ChAT) gene overexpressing mice (ChAT tg) show specific phenotypes including ischemic tolerance and the CNS stress tolerance. In the current study, we focused on molecular mechanisms responsible for systemic and localized anti-inflammatory phenotypes of ChAT tg. ChAT tg were resistant to systemic inflammation induced by lipopolysaccharides due to an attenuated cytokine response. In addition, ChAT tg, originally equipped with less reactive Kupffer cells, were refractory to brain cold injury, with decreased blood brain barrier (BBB) permeability and reduced inflammation. This is because ChAT tg brain endothelial cells expressed more claudin-5, and their astrocytes were less reactive, causing decreased hypertrophy. Moreover, reconstruction of the BBB integrity in vitro confirmed the consolidation of ChAT tg. ChAT tg were also resistant to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neuronal toxicity due to lower mortality rate and neuronal loss of substantia nigra. Additionally, ChAT tg subjected to MPTP showed attenuated BBB disruption, as evident from reduced sodium fluorescein levels in the brain parenchyma. The activated central cholinergic pathway of ChAT tg lead to anti-convulsive effects like vagus nerve stimulation. However, DSP-4, a noradrenergic neuron-selective neurotoxin against the CNS including the locus ceruleus, abrogated the beneficial phenotype and vagotomy attenuated expression of claudin-5, suggesting the link between the cholinergic pathway and BBB function. Altogether, these findings indicate that ChAT tg possess an anti-inflammatory response potential, associated with upregulated claudin-5, leading to the consolidation of BBB integrity. These characteristics protect ChAT tg against systemic and localized inflammatory pathological disorders, which targets the CNS.


Subject(s)
Blood-Brain Barrier/metabolism , Choline O-Acetyltransferase/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Acetylcholine/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Choline O-Acetyltransferase/physiology , Cholinergic Agents , Claudin-5/metabolism , Endothelial Cells/metabolism , Heart , Inflammation , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Permeability , Substantia Nigra/metabolism
4.
Cell Physiol Biochem ; 52(4): 922-934, 2019.
Article in English | MEDLINE | ID: mdl-30964609

ABSTRACT

BACKGROUND/AIMS: In a previous study, we reported that cardiomyocytes were equipped with non-neuronal cardiac cholinergic system (NNCCS) to synthesize acetylcholine (ACh), which is indispensable for maintaining the basic physiological cardiac functions. The aim of this study was to identify and characterize a pharmacological inducer of NNCCS. METHODS: To identify a pharmacological inducer of NNCCS, we screened several chemical compounds with chemical structures similar to the structure of S-nitroso-N-acetyl-DL-penicillamine (SNAP). Preliminary investigation revealed that SNAP is an inducer of non-neuronal ACh synthesis. We screened potential pharmacological inducers in H9c2 and HEK293 cells using western blot analysis, luciferase assay, and measurements of intracellular cGMP, NO2 and ACh levels. The effects of the screened compound on cardiac function of male C57BL6 mice were also evaluated using cardiac catheter system. RESULTS: Among the tested compounds, we selected S-nitroso-Npivaloyl-D-penicillamine (SNPiP), which gradually elevated the intracellular cGMP levels and nitric oxide (NO) levels in H9c2 and HEK293 cells. These elevated levels resulted in the gradual transactivation and translation of the choline acetyltransferase gene. Additionally, in vitro and in vivo SNPiP treatment elevated ACh levels for 72 h. SNPiP-treated mice upregulated their cardiac function without tachycardia but with enhanced diastolic function resulting in improved cardiac output. The effect of SNPiP was dependent on SNPiP nitroso group as verified by the ineffectiveness of N-pivaloyl-D-penicillamine (PiP), which lacks the nitroso group. CONCLUSION: SNPiP is identified to be one of the important pharmacological candidates for induction of NNCCS.


Subject(s)
Acetylcholine/biosynthesis , Cardiac Output/drug effects , Cyclic GMP/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide Donors , Non-Neuronal Cholinergic System/drug effects , Animals , HEK293 Cells , Humans , Male , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology
6.
J Cardiovasc Transl Res ; 10(4): 411-422, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28497301

ABSTRACT

Cardiomyocytes possess a non-neuronal cardiac cholinergic system (NNCCS) regulated by a positive feedback system; however, its other regulatory mechanisms remain to be elucidated, which include the epigenetic control or regulation by the female sex steroid, estrogen. Here, the NNCCS was shown to possess a circadian rhythm; its activity was upregulated in the light-off phase via histone acetyltransferase (HAT) activity and downregulated in the light-on phase. Disrupting the circadian rhythm altered the physiological choline acetyltransferase (ChAT) expression pattern. The NNCCS circadian rhythm may be regulated by miR-345, independently of HAT, causing decreased cardiac ChAT expression. Murine cardiac ChAT expression and ACh contents were increased more in female hearts than in male hearts. This upregulation was downregulated by treatment with the estrogen receptor antagonist tamoxifen, and in contrast, estrogen reciprocally regulated cardiac miR-345 expression. These results suggest that the NNCCS is regulated by the circadian rhythm and is affected by sexual dimorphism.


Subject(s)
Acetylcholine/metabolism , Choline O-Acetyltransferase/metabolism , Circadian Rhythm , Myocytes, Cardiac/enzymology , Periodicity , Animals , Cells, Cultured , Choline O-Acetyltransferase/genetics , Circadian Rhythm/drug effects , Epigenesis, Genetic , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Histone Acetyltransferases/metabolism , Humans , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , Ovariectomy , Photoperiod , Sex Characteristics , Sex Factors , Tamoxifen/pharmacology , Time Factors , Transcription, Genetic , Transfection
7.
Clin Sci (Lond) ; 130(21): 1913-28, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27528769

ABSTRACT

We previously developed cardiac ventricle-specific choline acetyltransferase (ChAT) gene-overexpressing transgenic mice (ChAT tgm), i.e. an in vivo model of the cardiac non-neuronal acetylcholine (NNA) system or non-neuronal cardiac cholinergic system (NNCCS). By using this murine model, we determined that this system was responsible for characteristics of resistance to ischaemia, or hypoxia, via the modulation of cellular energy metabolism and angiogenesis. In line with our previous study, neuronal ChAT-immunoreactivity in the ChAT tgm brains was not altered from that in the wild-type (WT) mice brains; in contrast, the ChAT tgm hearts were the organs with the highest expression of the ChAT transgene. ChAT tgm showed specific traits in a central nervous system (CNS) phenotype, including decreased response to restraint stress, less depressive-like and anxiety-like behaviours and anti-convulsive effects, all of which may benefit the heart. These phenotypes, induced by the activation of cardiac NNCCS, were dependent on the vagus nerve, because vagus nerve stimulation (VS) in WT mice also evoked phenotypes similar to those of ChAT tgm, which display higher vagus nerve discharge frequency; in contrast, lateral vagotomy attenuated these traits in ChAT tgm to levels observed in WT mice. Furthermore, ChAT tgm induced several biomarkers of VS responsible for anti-convulsive and anti-depressive-like effects. These results suggest that the augmentation of the NNCCS transduces an effective and beneficial signal to the afferent pathway, which mimics VS. Therefore, the present study supports our hypothesis that activation of the NNCCS modifies CNS to a more stress-resistant state through vagus nerve activity.


Subject(s)
Acetylcholine/metabolism , Central Nervous System/physiology , Heart Ventricles/metabolism , Heart/physiology , Animals , Central Nervous System/enzymology , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Heart Ventricles/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stress, Physiological , Vagus Nerve/enzymology , Vagus Nerve/metabolism
8.
Int Immunopharmacol ; 29(1): 181-4, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26072685

ABSTRACT

Ischemic preconditioning (IPC) renders the targeted organ resistant to prolonged ischemic insults, leading to organoprotection. Among several means to achieve IPC, we reported that remote ischemic preconditioning (RIPC) activates the non-neuronal cardiac cholinergic system (NNCCS) to accelerate de novo ACh synthesis in cardiomyocytes. In the current study, we aimed to optimize a specific protocol to most efficiently activate NNCCS using RIPC. In this study, we elucidated that the protocol with 3 min of ischemia repeated three times increased cardiac ChAT expression (139.2 ± 0.4%; P < 0.05) as well as ACh (14.2 ± 2.0× 10(-8) M; P< 0.05) and ATP content (2.13 ± 0.19 µmol/g tissue; P < 0.05) in the heart. Moreover, in the specific protocol, several characteristic responses against energy starvation and for obtaining adequate energy were observed; therefore, it is suggested that RIPC evokes a robust response by the heart to activate NNCCS through the modification of energy metabolism.


Subject(s)
Acetylcholine/metabolism , Adenosine Triphosphate/metabolism , Ischemic Preconditioning , Myocardium/metabolism , Animals , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Gene Expression Regulation , Hindlimb , Male , Mice , Mice, Inbred C57BL
9.
Int Immunopharmacol ; 29(1): 31-5, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25979761

ABSTRACT

We previously reported that satellite cells possess the ability to produce angiogenic factors, including fibroblast growth factor (FGF)-2 and vascular endothelial growth factor (VEGF) in vivo. However, whether C2C12 cells possess a non-neuronal cholinergic system (NNCS) or non-neuronal ACh (NNA) remains to be studied; therefore, we investigated the system using C2C12 cells and its regulatory mechanisms. C2C12 cells synthesized ACh, the level of which was comparable with that of cardiomyocytes, and the synthesis was augmented by the acetylcholinesterase inhibitor galantamine. The ChAT promoter activity was upregulated by nicotine or galantamine, partly through nicotinic receptors for both agents as well as through a non-nicotinic receptor pathway for galantamine. Further, VEGF secretion by C2C12 cells was also increased by nicotine or galantamine through nicotinic receptors as well as partly through non-nicotinic pathways in the case of galantamine. These results suggest that C2C12 cells are equipped with NNCS or NNA, which is positively regulated through nicotinic or non-nicotinic pathways, particularly in the case of galantamine. These results provide a novel concept that myogenic cells expressing NNA can be a therapeutic target for regulating angiogenic factor synthesis.


Subject(s)
Acetylcholine/metabolism , Cholinesterase Inhibitors/pharmacology , Galantamine/pharmacology , Receptors, Nicotinic/metabolism , Animals , Cell Line , Mice , Nicotine/pharmacology , Vascular Endothelial Growth Factor A/metabolism
10.
Cell Physiol Biochem ; 34(3): 781-9, 2014.
Article in English | MEDLINE | ID: mdl-25170772

ABSTRACT

BACKGROUND/AIMS: We previously suggested that a non-neuronal cholinergic system modulates energy metabolism through the mitochondria. However, the mechanisms responsible for making this system crucial remained undetermined. METHODS: In this study, we developed a fusion protein expression vector containing a luciferase gene fused to the folic acid receptor-α gene. RESULTS: This protein of the vector was confirmed to target the plasma membrane of transfected HEK293 cells, and vector-derived luciferase activities and ATP levels in viable cells were positively correlated (r = 0.599). Using this luciferase vector, choline acetyltransferase (ChAT)-expressing cells (i.e., cells with an activated non-neuronal cholinergic system) had increased cellular ATP levels. ChAT-expressing cells also had upregulated IGF-1R and Glut-1 protein expressions as well as increased glucose uptake. This activated non-neuronal cholinergic system with efficient glucose metabolism rendered cells resistant to serum depletion-induced cell death. CONCLUSION: Our results indicate that a non-neuronal cholinergic system is involved in sustaining ATP levels to render cells resistant to a nutrient-deficient environment.


Subject(s)
Adenosine Triphosphate/metabolism , Receptors, Cholinergic/metabolism , Cell Survival , Choline O-Acetyltransferase/metabolism , Culture Media , Culture Media, Serum-Free , HEK293 Cells , Humans
11.
Transl Res ; 164(1): 32-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24811002

ABSTRACT

We have recently identified that donepezil, an anti-Alzheimer drug, accelerates angiogenesis in a murine hindlimb ischemia (HLI) model. However, the precise mechanisms are yet to be fully elucidated, particularly whether the effects are derived from endothelial cells alone or from other nonvascular cells. Further investigation of the HLI model revealed that nicotine accelerated angiogenesis by activation of vascular endothelial cell growth factor (VEGF) synthesis through nicotinic receptors in myogenic cells, that is, satellite cells, in vivo and upregulated the expression of angiogenic factors, for example, VEGF and fibroblast growth factor 2, in vitro. As a result, nicotine prevented skeletal muscle from ischemia-induced muscle atrophy and upregulated myosin heavy chain expression in vitro. The in vivo anti-atrophy effect of nicotine on muscle was also observed in galantamine, another anti-Alzheimer drug, playing as an allosteric potentiating ligand. Such effects of nicotine were attenuated in α7 nicotinic receptor knockout mice. In contrast, PNU282987, an α7 nicotinic receptor agonist, comparably salvaged skeletal muscle, which was affected by HLI. These results suggest that cholinergic signals also target myogenic cells and have inhibiting roles in muscle loss by ischemia-induced muscle atrophy.


Subject(s)
Ischemia/metabolism , Muscular Atrophy/prevention & control , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Satellite Cells, Skeletal Muscle/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Cells, Cultured , Gene Expression Regulation/physiology , Hindlimb/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/blood supply , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...