Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Biosci Biotechnol Biochem ; 86(3): 351-361, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35015831

ABSTRACT

Tumor necrosis factor receptor 2 (TNFR2), a membrane-bound tumor necrosis factor receptor expressed by regulatory T cells (Tregs), participates in Treg proliferation. Although a specific TNFR2 pathway has been reported, the signaling mechanism has not been completely elucidated. This study sought to clarify TNFR2 signaling in human Tregs using amplicon sequencing and single-cell RNA sequencing to assess Tregs treated with a TNFR2 agonist antibody. Pathway enrichment analysis based on differentially expressed genes highlighted tumor necrosis factor α signaling via nuclear factor kappa B, interleukin-2 signal transducer and activator of transcription 5 signaling, interferon-γ response, and cell proliferation-related pathways in Tregs after TNFR2 activation. TNFR2-high Treg-focused analysis found that these pathways were fully activated in cancer Tregs, showing high TNFR2 expression. Collectively, these findings suggest that TNFR2 orchestrates multiple pathways in cancer Tregs, which could help cancer cells escape immune surveillance, making TNFR2 signaling a potential anticancer therapy target.


Subject(s)
Receptors, Tumor Necrosis Factor, Type II
3.
Biosci Biotechnol Biochem ; 85(3): 537-544, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33624776

ABSTRACT

Autoimmune responses to aquaporin 4 (AQP4) cause neuromyelitis optica (NMO); thus, specific immunotolerance to this self-antigen could represent a new NMO treatment. We generated the liposome-encapsulated AQP4 peptide 201-220 (p201-220) to induce immunotolerance. Liposomes were generated using phosphatidylserine and the polyglycidol species PG8MG. The in vivo tissue distribution of the liposomes was tested using an ex vivo imaging system. To confirm the antigen presentation capacity of PG8MG liposomes, dendritic cells were treated with PG8MG liposome-encapsulated AQP4 p201-220 (AQP4-PG8MG liposomes). Immunotolerance induction by AQP4-PG8MG liposomes was evaluated using the ex vivo cell proliferation of lymph node cells isolated from AQP4 p201-220-immunized AQP4-deficient mice. Fluorescent dye-labeled PG8MG liposomes were distributed to the lymph nodes. AQP4 p201-220 was presented on dendritic cells. AQP4-PG8MG liposomes were tended to suppress immune responses to AQP4 p201-220. Thus, the encapsulation of AQP4 peptides in PG8MG liposomes represents a new strategy for suppressing autoimmune responses to AQP4.


Subject(s)
Antigens/immunology , Aquaporin 4/immunology , Cell Proliferation , Histocompatibility Antigens Class II/immunology , Liposomes , Lymph Nodes/cytology , Peptides/immunology , Animals , Female , Mice , Mice, Inbred C57BL
4.
Biosci Biotechnol Biochem ; 83(6): 1000-1010, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30835622

ABSTRACT

Pulmonary hypertension (PH) is a life-threatening lung disease. PH with concomitant lung diseases, e.g., idiopathic pulmonary fibrosis, is associated with poor prognosis. Development of novel therapeutic vasodilators for treatment of these patients is a key imperative. We evaluated the efficacy of dual activation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) using an active, small-molecule phosphodiesterase (PDE4)/PDE5 dual inhibitor (Compound A). Compound A increased both cAMP and cGMP levels in WI-38 lung fibroblasts and suppressed the expressions of type-1 collagen α1 chain and fibronectin. Additionally, compound A reduced right ventricular weight/left ventricular weight+septal weight ratio, brain natriuretic peptide expression levels in right ventricle, C─C motif chemokine ligand 2 expression levels in lung, and plasma surfactant protein D. Our data indicate that dual activation of cAMP/cGMP pathways may be a novel treatment strategy for PH.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Inflammation/therapy , Lung/drug effects , Monocrotaline/toxicity , Phosphodiesterase 5 Inhibitors/therapeutic use , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Epithelium/injuries , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Lung/metabolism , Lung/pathology , Phosphodiesterase 5 Inhibitors/pharmacology , Rats, Wistar , Transforming Growth Factor beta/physiology
5.
Physiol Rep ; 6(12): e13753, 2018 06.
Article in English | MEDLINE | ID: mdl-29952109

ABSTRACT

Fibrosis of the lung constitutes a major clinical challenge and novel therapies are required to alleviate the associated morbidity and mortality. Investigating the antifibrotic efficacy of drugs that are already in clinical practice offers an efficient strategy to identify new therapies. The phosphodiesterase 4 (PDE4) inhibitors, approved for the treatment of chronic obstructive pulmonary disease, harbor therapeutic potential for pulmonary fibrosis by augmenting the activity of endogenous antifibrotic mediators that signal through cyclic AMP. In this study, we tested the efficacy of several PDE4 inhibitors including a novel compound (Compound 1) in a murine model of lung fibrosis that results from a targeted type II alveolar epithelial cell injury. We also compared the antifibrotic activity of PDE4 inhibition to the two therapies that are FDA-approved for idiopathic pulmonary fibrosis (pirfenidone and nintedanib). We found that both preventative (day 0-21) and therapeutic (day 11-21) dosing regimens of the PDE4 inhibitors significantly ameliorated the weight loss and lung collagen accumulation that are the sequelae of targeted epithelial cell damage. In a therapeutic protocol, the reduction in lung fibrosis with PDE4 inhibitor administration was equivalent to pirfenidone and nintedanib. Treatment with this class of drugs also resulted in a decrease in plasma surfactant protein D concentration, a reduction in the plasma levels of several chemokines implicated in lung fibrosis, and an in vitro inhibition of fibroblast profibrotic gene expression. These results motivate further investigation of PDE4 inhibition as a treatment for patients with fibrotic lung disease.


Subject(s)
Alveolar Epithelial Cells/pathology , Benzamides/therapeutic use , Isoquinolines/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Pulmonary Fibrosis/drug therapy , Aminopyridines/therapeutic use , Animals , Benzamides/administration & dosage , Benzamides/blood , Cells, Cultured , Chemokines/blood , Cyclic AMP/metabolism , Cyclopropanes/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Fibroblasts/metabolism , Humans , Isoquinolines/administration & dosage , Isoquinolines/blood , Mice, Inbred C57BL , Mice, Transgenic , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 4 Inhibitors/blood , Pulmonary Fibrosis/blood , Pulmonary Fibrosis/prevention & control , Pulmonary Surfactant-Associated Protein D/blood , Pyridines/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
6.
Chem Pharm Bull (Tokyo) ; 66(3): 270-276, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29311495

ABSTRACT

Pulmonary hypertension (PH) is a life-threatening lung disease. Despite the availability of several approved drugs, the development of a new treatment method is needed because of poor prognosis. Tissue selective drug delivery systems can avoid the adverse effects of current therapy and enhance efficacy. We evaluated the possibility of delivering drugs to the lungs of a PH rat model using fluorescence dye-labeled nanosized liposomes. To evaluate the tissue distribution following systemic exposure, fluorescent dye-labeled, 40-180 nm liposomes with and without polyethylene glycol (PEG) were intravenously administered to a monocrotaline-induced PH (MCT) rat model and tissue fluorescence was measured. Fluorescent dye-containing liposomes were intratracheally administered to the MCT model to evaluate the distribution of the liposome-encapsulated compound following local administration to reduce systemic exposure. The lung vascular permeability, plasma concentration of surfactant protein (SP)-D, lung reactive oxygen species (ROS) production, and macrophage marker gene cluster of differentiation (CD68) expression were measured. PEG and 80-nm liposome accumulation in the lung was elevated in the MCT model compared to that in normal rats. The intratracheally administered liposomes were delivered selectively to the lungs of the MCT model. The lung vascular permeability, plasma SP-D concentration, and CD68 expression were significantly elevated in the lungs of the MCT model, and were all significantly and positively correlated to liposome lung accumulation. Liposomes can accumulate in the lungs of an MCT model by enhancing vascular permeability by the inflammatory response. Therefore, drug encapsulation in liposomes could be an effective method of drug delivery in patients with PH.


Subject(s)
Fluorescent Dyes/metabolism , Hypertension, Pulmonary/metabolism , Liposomes/metabolism , Monocrotaline , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Drug Liberation , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/chemistry , Hypertension, Pulmonary/chemically induced , Liposomes/chemistry , Lung/drug effects , Lung/metabolism , Male , Optical Imaging , Particle Size , Permeability , Polyethylene Glycols/chemistry , Pulmonary Surfactant-Associated Protein D/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Surface Properties
7.
Biochem Biophys Res Commun ; 494(1-2): 188-193, 2017 12 09.
Article in English | MEDLINE | ID: mdl-29037813

ABSTRACT

Drug delivery systems maximize the efficacy of drugs by improving their pharmacokinetic profiles, pharmacodynamic effects, or both and reducing their adverse effects. One of the most advanced, clinically available formulations are liposome-encapsulated drugs. In this study, we aimed to determine if liposomes can selectively deliver compounds in gastrointestinal diseases. Initially, we evaluated the correlation between the diarrhea score and accumulation of fluorescence (FL)-labeled liposome using in vivo imaging systems in various disease states of an inflammatory bowel disease mouse model. The result showed that FL-labeled liposome accumulation and colon tissue weight, which reflect the disease state were highly and positively correlated. Then, to confirm the accumulation of liposomes at injured sites of the colon, we administered both FL-labeled liposomes and luminescence probes for detecting reactive oxygen species (ROS) to the mouse model. The imaging data showed that liposome accumulation tended to coincide with ROS detected sites and the correlation coefficient indicated a significantly positive correlation between liposome accumulation and ROS detection levels. Finally, we evaluated the involvement of macrophages in the uptake mechanism of the liposomes by analyzing the relationship between FL-labeled liposome accumulation and macrophage marker gene expression levels. The result showed that the expression of each macrophage marker gene and liposome accumulation showed a significant positive correlation. Therefore, the macrophages considerably contributed to the uptake mechanism of the liposomes. These data suggest that liposomes could be an attractive delivery tool for enhancing the accumulation of drug candidates through macrophages in injured colonic tissues. This approach is expected to provide new treatment options for patients with colitis.


Subject(s)
Colon/metabolism , Drug Delivery Systems , Inflammatory Bowel Diseases/drug therapy , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, Differentiation/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Colon/injuries , Disease Models, Animal , Female , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/pharmacokinetics , Genetic Markers , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Liposomes , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nanomedicine , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class A/genetics , T-Lymphocytes/immunology
8.
FASEB J ; 31(12): 5307-5320, 2017 12.
Article in English | MEDLINE | ID: mdl-28798156

ABSTRACT

Duchenne muscular dystrophy (DMD) is the most common inherited muscular dystrophy. Patients experience DMD in their 20s from cardiac or respiratory failure related to progressive muscle wasting. Currently, the only treatments for the symptoms of DMD are available. Muscle fibrosis, a DMD feature, leads to reduced muscle function and muscle mass, and hampers pharmaceutical therapeutic efficacy. Although antifibrotic agents may be useful, none is currently approved. Phosphodiesterase 4 (PDE4) inhibitors have exhibited antifibrotic effects in human and animal models. In this study, we showed beneficial effects of the PDE4 inhibitor piclamilast in the DMD mdx mouse. Piclamilast reduced the mRNA level of profibrotic genes, including collagen 1A1, in the gastrocnemius and diaphragm, in the mdx mouse, and significantly reduced the Sirius red staining area. The PDE5 inhibitors sildenafil and tadalafil ameliorated functional muscle ischemia in boys with DMD, and sildenafil reversed cardiac dysfunction in the mdx mouse. Single-treatment piclamilast or sildenafil showed similar antifibrotic effects on the gastrocnemius; combination therapy showed a potent antifibrotic effect, and piclamilast and combination therapy increased peroxisome proliferator-activated receptor γ coactivator-1α mRNA in mouse gastrocnemius. In summary, we confirmed that piclamilast has significant antifibrotic effects in mdx mouse muscle and is a potential treatment for muscle fibrosis in DMD.-Nio, Y., Tanaka, M., Hirozane, Y., Muraki, Y., Okawara, M., Hazama, M., Matsuo, T. Phosphodiesterase 4 inhibitor and phosphodiesterase 5 inhibitor combination therapy has antifibrotic and anti-inflammatory effects in mdx mice with Duchenne muscular dystrophy.


Subject(s)
Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Benzamides/therapeutic use , Fibrosis/drug therapy , Fibrosis/enzymology , Fibrosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/metabolism , PPAR gamma/genetics , Pyridines/therapeutic use , RNA, Messenger/genetics , Sildenafil Citrate/therapeutic use
9.
Bioorg Med Chem ; 25(15): 4175-4193, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28642028

ABSTRACT

Somatostatin receptor subtype 5 (SSTR5) has emerged as a novel attractive drug target for type 2 diabetes mellitus. Starting from N-benzyl azetidine derivatives 1 and 2 as in-house hit compounds, we explored the introduction of a carboxyl group into the terminal benzene of 1 to enhance SSTR5 antagonistic activity by the combination of the substituents at the 3-position of the isoxazoline. Incorporation of a carboxyl group at the 4-position of the benzene ring resulted in a significant enhancement in potency, however, the 4-benzoic acid derivative 10c exhibited moderate human ether-a-go-go related gene (hERG) inhibitory activity. A subsequent optimization study revealed that replacement of the 4-benzoic acid with an isonipecotic acid dramatically reduced hERG inhibition (5.6% inhibition at 30µM) by eliminating π-related interaction with hERG K+ channel, which resulted in the identification of 1-(2-((2,6-diethoxy-4'-fluorobiphenyl-4-yl)methyl)-5-oxa-2,6-diazaspiro[3.4]oct-6-en-7-yl)piperidin-4-carboxylic acid 25a (hSSTR5/mSSTR5 IC50=9.6/57nM). Oral administration of 25a in high-fat diet fed C57BL/6J mice augmented insulin secretion in a glucose-dependent manner and lowered blood glucose concentration.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Animals , CHO Cells , Carbon-13 Magnetic Resonance Spectroscopy , Cricetulus , Drug Discovery , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Proton Magnetic Resonance Spectroscopy
10.
Biochem Biophys Res Commun ; 486(3): 632-638, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28322783

ABSTRACT

Endoplasmic reticulum (ER) stress caused by accumulation of misfolded proteins is observed in several kinds of diseases. Since ER stress is reported to be involved in the progression of non-alcoholic steatohepatitis (NASH), highly sensitive and simple measurement methods are required for research into developing novel therapy for NASH. To investigate the involvement of ER stress in NASH pathogenesis in a mouse model, an assay for liver ER stress was developed using ER stress activated indicator-luciferase (ERAI-Luc) mice. To establish the assay method for detection of ER stress in the liver, tunicamycin (TM) (0.3 mg/kg i. p.) was administered to ERAI-Luc mice, and the luciferase activity was measured in ex vivo and in vivo. To evaluate ER stress in the NASH model, ERAI-Luc mice were fed a modified choline-deficient l-amino acid-defined (mCDAA) diet for 14 weeks. After measurement of ER stress by luminescence imaging, levels of liver lipids and pro-fibrotic and pro-inflammatory gene expression were measured as NASH-related indexes. In non-invasive whole-body imaging, TM elevated luciferase activity in the liver, induced by activation of ER stress. The highest luminescence in the liver was confirmed by ex vivo imaging of isolated tissues. In parallel with progression of NASH, elevated luminescence induced by ER stress in liver was observed in mCDAA diet-fed ERAI-Luc mice. Luciferase activity was significantly and positively correlated to levels of triglyceride and free cholesterol in the liver, as well as to the mRNA expression of type 1 collagen α1 chain and tumor necrosis factor α. These data indicated that the use of ERAI-Luc mice was effective in the detection of ER stress in the liver. Moreover, the NASH model using ERAI-Luc mice can be a useful tool to clarify the role of ER stress in pathogenesis of NASH and to evaluate effects of drugs targeted against ER stress.


Subject(s)
Choline Deficiency/genetics , Collagen Type I/genetics , Endoplasmic Reticulum Stress/genetics , Food, Formulated/adverse effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Biological Assay , Cholesterol/metabolism , Choline Deficiency/etiology , Choline Deficiency/metabolism , Choline Deficiency/pathology , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation , Genes, Reporter , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tunicamycin/pharmacology
11.
J Neurophysiol ; 96(1): 284-98, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16611835

ABSTRACT

We reported elsewhere that orexin neurons are directly hyperpolarized by noradrenaline (NA) and dopamine. In the present study, we show that NA, dopamine, and adrenaline all directly hyperpolarized orexin neurons. This response was inhibited by the alpha2 adrenergic receptor (alpha2-AR) antagonist, idazoxan or BRL44408, and was mimicked by the alpha2-AR-selective agonist, UK14304. A low concentration of Ba2+ inhibited NA-induced hyperpolarization, which suggests that activation of G protein coupled inward rectifier potassium channels is involved in the response. In the presence of a high concentration of idazoxan, NA induced depolarization or inward current. This response was inhibited by alpha1-AR antagonist, prazosin, which suggests the existence of alpha1-ARs on the orexin neurons along with alpha2-AR. We also examined the effects of NA on glutamatergic and GABAergic synaptic transmission. NA application dramatically increased the frequency and amplitude of spontaneous inhibitory synaptic currents (sIPSCs) and inhibited excitatory synaptic currents (sEPSCs) in orexin neurons; however, NA decreased the frequency of miniature EPSCs (mEPSCs) and IPSCs and the amplitude of evoked EPSCs and IPSCs through the alpha2-AR, because the NA response on mPSCs was inhibited by idazoxan. These results suggest that the NA-induced increase in sIPSC frequency and amplitude is mediated via alpha1-ARs on the somata of GABAergic neurons that innervate the orexin neurons. Calcium imaging using orexin/YC2.1 transgenic mouse brain revealed that NA-induced inhibition of orexin neurons is not altered by sleep deprivation or circadian time in mice. The evidence presented here revealed that orexin neurons are regulated by catecholamines in a complex manner.


Subject(s)
Catecholamines/physiology , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/physiology , Neurons/chemistry , Neurons/physiology , Neuropeptides/analysis , Neuropeptides/physiology , Action Potentials/drug effects , Action Potentials/physiology , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Calcium/physiology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Idazoxan/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Neurons/drug effects , Norepinephrine/physiology , Orexin Receptors , Orexins , Receptors, Adrenergic, alpha-1/analysis , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, alpha-2/analysis , Receptors, Adrenergic, alpha-2/physiology , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Sleep Deprivation/physiopathology , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/physiology
12.
J Neurosci ; 25(32): 7459-69, 2005 Aug 10.
Article in English | MEDLINE | ID: mdl-16093397

ABSTRACT

Orexin A and B are neuropeptides implicated in the regulation of sleep/wakefulness and energy homeostasis. The regulatory mechanism of the activity of orexin neurons is not precisely understood. Using transgenic mice in which orexin neurons specifically express yellow cameleon 2.1, we screened for factors that affect the activity of orexin neurons (a total of 21 peptides and six other factors were examined) and found that a sulfated octapeptide form of cholecystokinin (CCK-8S), neurotensin, oxytocin, and vasopressin activate orexin neurons. The mechanisms that underlie CCK-8S-induced activation of orexin neurons were studied by both calcium imaging and slice patch-clamp recording. CCK-8S induced inward current in the orexin neurons. The CCKA receptor antagonist lorglumide inhibited CCK-8S-induced activation of orexin neurons, whereas the CCKB receptor agonists CCK-4 (a tetrapeptide form of cholecystokinin) and nonsulfated CCK-8 had little effect. The CCK-8S-induced increase in intracellular calcium concentration was eliminated by removing extracellular calcium but not by an addition of thapsigargin. Nifedipine, omega-conotoxin, omega-agatoxin, 4-ethylphenylamino-1,2-dimethyl-6-methylaminopyrimidinium chloride, and SNX-482 had little effect, but La3+, Gd3+, and 2-aminoethoxydiphenylborate inhibited CCK-8S-induced calcium influx. Additionally, the CCK-8S-induced inward current was dramatically enhanced in the calcium-free solution and was inhibited by the cation channel blocker SKF96365, suggesting an involvement of extracellular calcium-sensitive cation channels. CCK-8S did not induce an increase in intracellular calcium concentration when membrane potential was clamped at -60 mV, suggesting that the calcium increase is induced by depolarization. The evidence presented here expands our understanding of the regulation of orexin neurons and the physiological role of CCK in the CNS.


Subject(s)
Cholecystokinin/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Neuropeptides/metabolism , Receptor, Cholecystokinin A/physiology , Animals , Calcium/metabolism , Electric Conductivity , Excitatory Postsynaptic Potentials/drug effects , Extracellular Fluid/metabolism , Female , Fluorescent Antibody Technique , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Neural Inhibition/physiology , Neurons/drug effects , Neurons/metabolism , Orexin Receptors , Orexins , Patch-Clamp Techniques , Receptor, Cholecystokinin A/metabolism , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sincalide/analogs & derivatives , Sincalide/pharmacology , Synaptic Transmission/drug effects
13.
Neuron ; 46(2): 297-308, 2005 Apr 21.
Article in English | MEDLINE | ID: mdl-15848807

ABSTRACT

The finding of orexin/hypocretin deficiency in narcolepsy patients suggests that this hypothalamic neuropeptide plays a crucial role in regulating sleep/wakefulness states. However, very little is known about the synaptic input of orexin/hypocretin-producing neurons (orexin neurons). We applied a transgenic method to map upstream neuronal populations that have synaptic connections to orexin neurons and revealed that orexin neurons receive input from several brain areas. These include the amygdala, basal forebrain cholinergic neurons, GABAergic neurons in the preoptic area, and serotonergic neurons in the median/paramedian raphe nuclei. Monoamine-containing groups that are innervated by orexin neurons do not receive reciprocal connections, while cholinergic neurons in the basal forebrain have reciprocal connections, which might be important for consolidating wakefulness. Electrophysiological study showed that carbachol excites almost one-third of orexin neurons and inhibits a small population of orexin neurons. These neuroanatomical findings provide important insights into the neural pathways that regulate sleep/wakefulness states.


Subject(s)
Hypothalamus/anatomy & histology , Intracellular Signaling Peptides and Proteins/metabolism , Neural Pathways/anatomy & histology , Neurons/cytology , Neuropeptides/metabolism , Animals , Brain Stem/anatomy & histology , Brain Stem/drug effects , Brain Stem/ultrastructure , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Green Fluorescent Proteins/genetics , Humans , Hypothalamus/drug effects , Hypothalamus/ultrastructure , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Neural Pathways/drug effects , Neurons/physiology , Orexins , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Tetrodotoxin/genetics , Wakefulness/physiology
14.
J Neurosci ; 24(32): 7159-66, 2004 Aug 11.
Article in English | MEDLINE | ID: mdl-15306649

ABSTRACT

Both orexin and serotonin (5-HT) have important roles in the regulation of sleep-wakefulness, as well as in feeding behavior. We examined the effects of 5-HT on orexin/hypocretin neurons, using hypothalamic slices prepared from orexin/enhanced green fluorescent protein (EGFP) transgenic mice in which EGFP is expressed exclusively in orexin neurons. Patch-clamp recording from EGFP-expressing cells showed that 5-HT hyperpolarized all orexin neurons in a concentration-dependent manner. The response was inhibited by the 5-HT1A receptor antagonist WAY100635. A 5-HT1A receptor agonist, 8-hydroxy-2-(dl-N-propyl-amino)tetralin, also evoked hyperpolarization on orexin neurons with potency comparable with 5-HT. A low concentration of Ba2+ (30 microM) inhibited 5-HT-induced hyperpolarization. Single-channel recording revealed that the conductance of 5-HT-induced channel activity was 33.8 pS, which is in good agreement with that of the G-protein-coupled inward rectifier potassium channel (GIRK). Moreover, 5-HT1A receptor-like immunoreactivity was observed on orexin neurons, and 5-HT transporter immunoreactive nerve endings are in close apposition to orexin neurons. Intracerebroventricular injection of the 5-HT1A receptor-selective antagonist WAY100635 (100 ng) increased locomotor activity during the latter half of dark phase in wild-type mice but not in orexin/ataxin-3 mice in which orexin neurons are specifically ablated, suggesting that activation of orexin neurons is necessary for the WAY100635-induced increase in locomotor activity. These results indicate that 5-HT hyperpolarizes orexin neurons through the 5-HT1A receptor and subsequent activation of the GIRK and that this inhibitory serotonergic input to the orexin neurons is likely to be important for the physiological regulation of this neuropeptide system.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Neuropeptides/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Animals , Ataxin-3 , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , In Vitro Techniques , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/genetics , Nuclear Proteins , Orexin Receptors , Orexins , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/agonists , Receptor, Serotonin, 5-HT1A/biosynthesis , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Repressor Proteins , Serotonin/pharmacology , Serotonin Antagonists/administration & dosage , Serotonin Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins , Serotonin Receptor Agonists/pharmacology , Tetrodotoxin/pharmacology , Transcription Factors
15.
Biochem Biophys Res Commun ; 303(1): 120-9, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12646175

ABSTRACT

Orexins are a pair of neuropeptides implicated in energy homeostasis and arousal. Here we characterize the electrophysiological properties of orexin neurons using slice preparations from transgenic mice in which orexin neurons specifically express green fluorescent protein. Orexin neurons showed high frequency firing with little adaptation by injecting a positive current. The hyperpolarization-activated current was observed in orexin neurons by a negative current injection. The neurotransmitters, which were implicated in sleep/wake regulation, affected the activity of orexin neurons; noradrenaline and serotonin hyperpolarized, while carbachol depolarized orexin neurons in either the presence or absence of tetrodotoxin. It has been reported that orexins directly or indirectly activate the nuclei that are the origin of the neurons containing these neurotransmitters. Our data suggest that orexin neurons have reciprocal neural circuitries between these nuclei for either a positive or negative feedback loop and orchestrate the activity of these neurons to regulate the vigilance states.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Neurons/metabolism , Neuropeptides/metabolism , Acetylcholine/metabolism , Action Potentials , Animals , Electrophysiology , Female , Green Fluorescent Proteins , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , N-Methylaspartate/pharmacology , Neurotransmitter Agents/metabolism , Norepinephrine/metabolism , Orexins , Patch-Clamp Techniques , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...