Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Acta Diabetol ; 61(6): 705-714, 2024 Jun.
Article En | MEDLINE | ID: mdl-38400938

AIMS: Several studies have reported dietary microorganisms' beneficial effects on human health. We aimed to detect the potential association between dietary live microbe intake and diabetic kidney disease (DKD) in patients with type 2 diabetes mellitus (T2DM) through a cross-sectional analysis of the National Health and Nutrition Examination Survey from 1999 to 2018. METHODS: According to the Sanders classification system of dietary live microbes, the study participants were divided into three groups: low, medium, and high live microbe groups. In patients with T2DM, DKD was assessed by glomerular filtration rate (< 60 mL/min/1.73 m2 using the Chronic Kidney Disease Epidemiology Collaboration algorithm), proteinuria (urinary albumin to creatinine ratio ≥ 30 mg/g), or both. Weighted univariate and multivariate logistic regression and subgroup analyses were conducted to investigate the independent association between dietary live microbe and DKD. RESULTS: The study included 3836 participants, of whom 1467 (38.24%) had DKD for the diagnosis. Our study demonstrated that participants in the high dietary live microbe group were more likely to be older, female, non-Hispanic White, have higher education levels, have a lower prevalence of smoking, have a high poverty-income ratio, have higher energy intake, lower haemoglobin (HbA1c) and serum creatinine levels, and lower risk of progression. After adjustment for covariates, patients in the high dietary live microbe group had a low prevalence of DKD, whereas no significant association with DKD was found between the medium and low dietary live microbe groups. No statistically significant interaction was observed in all subgroup analyses except for HbA1c (p for interaction < 0.05). CONCLUSIONS: Our results indicate that high dietary live microbe intake was associated with a low DKD prevalence.


Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Nutrition Surveys , Humans , Female , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Male , Cross-Sectional Studies , Middle Aged , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/etiology , Aged , Adult , Diet/statistics & numerical data , United States/epidemiology , Glomerular Filtration Rate
2.
Biochem Biophys Res Commun ; 501(4): 827-832, 2018 07 02.
Article En | MEDLINE | ID: mdl-29654764

Recent study has showed that Ginsenoside Rg1, the mian active compound of Panax ginseng, could ameliorate oxidative stress and myocardial apoptosis in diabetes mellitus. However, the roles and mechanisms of Rg1 in proliferative diabetic retinopathy (PDR) are still unclear. In the present study, we aimed to investigate the effects of Rg1 on mesenchymal activation of high-glucose (HG) cultured müller cells. High glucose conditions up-regulate MMP-2, MMP-9 and down-regulate TIMP-2, and promote mesenchymal activation in Müller cells. And Rg1 inhibits the HG-induced mesenchymal activation and HG-increased MMP-2 and MMP-9 and HG-decreased TIMP-2 in Müller cells. HG up-regulates Zeb1 and lncRNA RP11-982M15.8, and down-regulates miR-2113, and Rg1 inhibits these effects of HG. Both inhibition of miR-2113 and over-expression of RP11-982M15.8 significantly restored the HG induced mesenchymal activasion. Taken together, our findings suggested that Rg1 inhibited HG-induced mesenchymal activation and fibrosis via regulating miR-2113/RP11-982M15.8/Zeb1 pathway.


Ginsenosides/pharmacology , Glucose/toxicity , Mesoderm/metabolism , Mesoderm/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Fibrosis , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9 , Mesoderm/drug effects , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
3.
Neuropeptides ; 67: 36-44, 2018 Feb.
Article En | MEDLINE | ID: mdl-29195839

Neuropeptide S (NPS), the endogenous neuropeptide ligand of NPSR, has been reported to regulate anxiety-related behavior involved in multiple brain regions, including amygdale, locus coeruleus and Barrington's nucleus. However, little research has been conducted on the anxiolytic-like behaviors of NPS on the hypothalamus, which was an important area in defensive behavior. Here, we investigated a role of hypothalamus in anxiolytic-like behaviors of NPS. We found that NPSR protein of mouse distributed mainly in the ventromedial hypothalamus (VMH). And in the single prolonged stress model (SPS), the results showed that NPS mRNA of the mice exposed to SPS was significantly higher than control, while NPSR mRNA was remarkable lower than control in hypothalamus. Further studies found that NPS intra-VMH infusion dose-dependently (1, 10 and 100pmol) induced anxiolytic effects, using elevated plus maze and open field tests. These anxiolytic effects could be blocked by NPSR antagonist (SHA68), but not by picrotoxin (a GABAA receptor antagonist) and sacolfen (a GABAB receptor antagonist). Meanwhile, our data showed that the expression of c-Fos was significantly increased in VMH after NPS delivered into the lateral ventricles. These results cast a new light on the hypothalamic nucleus in the anxiolytic-like effect of NPS system.


Anti-Anxiety Agents/pharmacology , Hypothalamus/drug effects , Motor Activity/drug effects , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , Animals , Male , Maze Learning/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Receptors, Neuropeptide/drug effects
4.
Brain Res ; 1629: 210-20, 2015 Dec 10.
Article En | MEDLINE | ID: mdl-26505914

A growing body of evidence suggests that the agglomeration of amyloid-ß (Aß) may be a trigger for Alzheimer׳s disease (AD). Central infusion of Aß42 can lead to memory impairment in mice. Inhibiting the aggregation of Aß has been considered a therapeutic strategy for AD. Endomorphin-1 (EM-1), an endogenous agonist of µ-opioid receptors, has been shown to inhibit the aggregation of Aß in vitro. In the present study, we investigated whether EM-1 could alleviate the memory-impairing effects of Aß42 in mice using novel object recognition (NOR) and object location recognition (OLR) tasks. We showed that co-administration of EM-1 was able to ameliorate Aß42-induced amnesia in the lateral ventricle and the hippocampus, and these effects could not be inhibited by naloxone, an antagonist of µ-opioid receptors. Infusion of EM-1 or naloxone separately into the lateral ventricle had no influence on memory in the tasks. These results suggested that EM-1 might be effective as a drug for AD preventative treatment by inhibiting Aß aggregation directly as a molecular modifier.


Amyloid beta-Peptides/toxicity , Analgesics, Opioid/administration & dosage , Memory Disorders/chemically induced , Memory Disorders/prevention & control , Oligopeptides/administration & dosage , Peptide Fragments/toxicity , Recognition, Psychology/drug effects , Amyloid beta-Peptides/administration & dosage , Animals , Infusions, Intraventricular , Male , Mice , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Peptide Fragments/administration & dosage , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/physiology , Recognition, Psychology/physiology
5.
Neurobiol Learn Mem ; 107: 32-6, 2014 Jan.
Article En | MEDLINE | ID: mdl-24211255

The noradrenergic activity in the basolateral amygdala (BLA) was reported to be involved in the regulation of object recognition memory. As the BLA expresses high density of receptors for Neuropeptide S (NPS), we investigated whether the BLA is involved in mediating NPS's effects on object recognition memory consolidation and whether such effects require noradrenergic activity. Intracerebroventricular infusion of NPS (1nmol) post training facilitated 24-h memory in a mouse novel object recognition task. The memory-enhancing effect of NPS could be blocked by the ß-adrenoceptor antagonist propranolol. Furthermore, post-training intra-BLA infusions of NPS (0.5nmol/side) improved 24-h memory for objects, which was impaired by co-administration of propranolol (0.5µg/side). Taken together, these results indicate that NPS interacts with the BLA noradrenergic system in improving object recognition memory during consolidation.


Amygdala/physiology , Neuropeptides/pharmacology , Recognition, Psychology/physiology , Adrenergic beta-Antagonists/pharmacology , Amygdala/drug effects , Animals , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Mice , Propranolol/pharmacology , Recognition, Psychology/drug effects
6.
Neuropharmacology ; 70: 261-7, 2013 Jul.
Article En | MEDLINE | ID: mdl-23454528

Neuropeptide S (NPS), the endogenous ligand of NPSR, has been shown to promote arousal and anxiolytic-like effects. According to the predominant distribution of NPSR in brain tissues associated with learning and memory, NPS has been reported to modulate cognitive function in rodents. Here, we investigated the role of NPS in memory formation, and determined whether NPS could mitigate memory impairment induced by selective N-methyl-D-aspartate receptor antagonist MK801, muscarinic cholinergic receptor antagonist scopolamine or Aß1₋42 in mice, using novel object and object location recognition tasks. Intracerebroventricular (i.c.v.) injection of 1 nmol NPS 5 min after training not only facilitated object recognition memory formation, but also prolonged memory retention in both tasks. The improvement of object recognition memory induced by NPS could be blocked by the selective NPSR antagonist SHA 68, indicating pharmacological specificity. Then, we found that i.c.v. injection of NPS reversed memory disruption induced by MK801, scopolamine or Aß1₋42 in both tasks. In summary, our results indicate that NPS facilitates memory formation and prolongs the retention of memory through activation of the NPSR, and mitigates amnesia induced by blockage of glutamatergic or cholinergic system or by Aß1₋42, suggesting that NPS/NPSR system may be a new target for enhancing memory and treating amnesia.


Amyloid beta-Peptides/antagonists & inhibitors , Dizocilpine Maleate/antagonists & inhibitors , Memory Disorders/drug therapy , Memory/drug effects , Neuropeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Recognition, Psychology/drug effects , Scopolamine/antagonists & inhibitors , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/pharmacology , Animals , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Infusions, Intraventricular , Male , Memory Disorders/chemically induced , Mice , Neuropeptides/administration & dosage , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Receptors, Neuropeptide/agonists , Scopolamine/administration & dosage , Scopolamine/pharmacology
7.
Brain Res ; 1470: 59-68, 2012 Aug 27.
Article En | MEDLINE | ID: mdl-22750583

Our previous reports indicated that bis(propyl)-cognitin (B3C) and bis(heptyl)-cognitin (B7C), as novel dimers derived from tacrine, may be potential multifunctional drugs for treating Alzheimer's disease. There is little knowledge on the cognitive function of B3C while B7C appeared to reverse learning and memory impairments. In this study, for the first time, we evaluated the anti-amnesic effects of B3C and B7C on learning and memory deficits induced by scopolamine using both Morris water maze and novel object recognition tasks in mice. Under the same experimental condition, the anti-amnesic effect of tacrine was also compared. Briefly, in both tasks, scopolamine (0.1-0.6 mg/kg, ip) dose-dependently impaired learning and memory functions. B3C (1.5-2.5 µmol/kg), B7C (0.4-0.6 µmol/kg) or tacrine (8-12 µmol/kg), each administered ip, dose-dependently mitigated scopolamine-induced learning and memory impairments in both tasks. Our present results show, for the first time, that B3C and B7C reverse cognitive impairment resulted from scopolamine in both water maze and object recognition tasks; and under the same condition, the relative potency of B3C and B7C to improve cognitive capacity was 5-20 folds over that of tacrine. These novel in vivo findings further demonstrate that both B3C and B7C may potentially be developed as Alzheimer's therapeutic drugs for different severities of neurodegenerations.


Cholinesterase Inhibitors/therapeutic use , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Recognition, Psychology/drug effects , Tacrine/therapeutic use , Analysis of Variance , Animals , Cholinergic Antagonists/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Learning Disabilities/chemically induced , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Mice , Mice, Inbred Strains , Reaction Time/drug effects , Retention, Psychology/drug effects , Scopolamine/toxicity
8.
Peptides ; 31(12): 2259-63, 2010 Dec.
Article En | MEDLINE | ID: mdl-20800637

Neuropeptide S (NPS), the endogenous ligand of NPS receptor (NPSR), can regulate a variety of biological functions, including arousal, anxiety, locomotion, memory and drug abuse. Previous studies have shown that central NPS inhibited food intake in rats and chicks. In the present study, we investigated the role of central NPS on food intake in fasted mice, and detected the underlying mechanism(s) by using NPSR antagonist [D-Val(5)]NPS and Corticotropin-Releasing Factor 1 (CRF1) Receptor antagonist NBI-27914. The present results indicated that intracerebroventricular injection of NPS (0.001-0.1 nmol) dose-dependently inhibited food intake in fasted mice. The anorectic effect of NPS reached the maximum at the dose of 0.1 nmol, which could be antagonized by co-injection of 10 nmol NPSR antagonist [D-Val(5)]NPS. Furthermore, CRF1 receptor antagonist NBI-27914 at the dose of 2 µg antagonized the hyperlocomotor action of NPS, but did not affect the role of NPS on food intake. In conclusion, our results demonstrated central NPS inhibited food intake in fasted mice, mediated by its cognate NPSR, but not by CRF1 receptor.


Eating/drug effects , Neuropeptides/pharmacology , Receptors, Neuropeptide/metabolism , Aniline Compounds/pharmacology , Animals , Male , Mice , Neuropeptides/chemical synthesis , Neuropeptides/chemistry , Pyrimidines/pharmacology , Rats , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
9.
Peptides ; 31(10): 1878-83, 2010 Oct.
Article En | MEDLINE | ID: mdl-20603169

Neuropeptide S (NPS), a recently discovered bioactive peptide, was reported to regulate arousal, anxiety, locomotion, feeding behaviors, memory, and drug addiction. NPS receptor (NPSR) mRNA was found in several brain regions related to descending control system of pain, including the periaqueductal gray (PAG). Our previous study had shown that NPS could produce antinociception in mice. The present study was designed to evaluate whether NPS may produce antinociceptive effect observed in the mouse formalin test, a model of inflammatory pain. NPS (0.1-100 pmol) administrated intracerebroventricularly (i.c.v.) dose-dependently attenuated both first-phase and second-phase nociceptive behaviors induced by paw formalin injection. NPS (10 pmol, i.c.v.)-elicited antinociceptive effect was counteracted by co-injection with 1000 and 10,000 pmol [D-Val(5)]NPS, which alone induced neither hyperalgesia nor antinociception. The antinociception induced by NPS (10 pmol, i.c.v.) was not affected by naloxone (i.p., 10 mg/kg) and naloxone alone had no effect in the formalin test. In addition, compared to the saline (i.c.v.) treated group, NPS (10 pmol, i.c.v.) treated group increased c-Fos protein expression in nearly all subdivisions of the PAG in the formalin-injected mice. The above results revealed that NPS could produce antinociception in the formalin test through NPSR, which may be involved in the activation of PAG, suggesting that NPS-NPSR system may be a potential target for developing new analgesic drugs.


Analgesics , Neuropeptides , Pain Measurement/drug effects , Pain/drug therapy , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Male , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Periaqueductal Gray/cytology , Periaqueductal Gray/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
10.
Peptides ; 30(7): 1313-7, 2009 Jul.
Article En | MEDLINE | ID: mdl-19540430

Neuropeptide S (NPS), the endogenous ligand of NPS receptor (NPSR), regulates many biological functions, including arousal, anxiety, locomotion and food intake. NPSR mRNA is expressed in several regions of central autonomic network through which the brain controls visceromotor and other responses essential for survival. However, the role of NPS/NPSR system in regulating gastrointestinal motor is still unknown. Here, we studied the effects of NPS on distal colonic transit in mice. Intracerebroventricular (i.c.v.) injection of NPS (1-1000 pmol) inhibited fecal pellet output and bead expulsion in a dose-dependent manner. However, intraperitoneal injection of NPS (1000 and 10000 pmol) did not affect fecal pellet output and bead expulsion. In vitro, NPS (0.1-10 microM) also did not modulate distal colonic contractions. Furthermore, i.c.v. co-administration of [D-Val(5)]NPS, a pure and potent NPSR antagonist, dose-dependently antagonized the inhibitory effects of NPS on fecal pellet output and bead expulsion. In conclusion, our results firstly indicate that central NPS inhibits distal colonic transit through the activation of central NPSR, which implicate that NPS/NPSR system might be a new target to treat function disorder of distal colon.


Colon/drug effects , Gastrointestinal Motility/drug effects , Gastrointestinal Transit/drug effects , Neuropeptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Colon/physiology , In Vitro Techniques , Injections, Intraperitoneal , Injections, Intraventricular , Male , Mice , Receptors, G-Protein-Coupled/antagonists & inhibitors
11.
Neurosci Lett ; 455(1): 74-7, 2009 May 08.
Article En | MEDLINE | ID: mdl-19429110

Neuropeptide S (NPS) is a recently discovered peptide shown to be involved in regulating arousal and anxiety. NPS receptor (NPSR) mRNA is expressed significantly in the major input and output regions of hippocampal formation, which are critical in the modulation of learning and memory. However, the role of NPS/NPSR system in regulating of learning and memory is still unknown. Here, we use the Morris water maze (MWM) to determine the effects of NPS on spatial learning and memory following intracerebroventricular (i.c.v.) injection in mice. Our data show that i.c.v. injection of NPS facilitates spatial memory in the MWM without significant alteration of latency to the target and swimming speed. Furthermore, NPS (i.c.v.) mitigates spatial memory impairment induced by the selective N-methyl-d-aspartate receptor antagonist MK801. Taken together, our results firstly demonstrate that NPS facilitates spatial memory and mitigates MK801-induced spatial memory impairment in mice.


Dizocilpine Maleate/pharmacology , Memory/drug effects , Neuropeptides/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Injections, Intraventricular , Male , Maze Learning/drug effects , Mice , Neuropeptides/pharmacology
12.
Brain Res ; 1271: 36-48, 2009 May 19.
Article En | MEDLINE | ID: mdl-19332041

The mas-related genes (Mrgs, also known as sensory neuron-specific receptors, SNSRs) are specifically expressed in small diameter sensory neurons in the trigeminal and dorsal root ganglia, suggesting an important role of the receptors in pain transmission. The present study aimed to investigate the underlying mechanism of the nociceptive effects after activation of MrgC, and the interaction between MrgC and N/OFQ-NOP receptor system in modulation of nociception in mice. Intrathecal (i.t.) administration of [Tyr(6)] gamma2-MSH(6-12), the most potent agonist for MrgC receptor, produced a significant hyperalgesic response as assayed by tail withdrawal test and a series of characteristic nociceptive responses, including biting, licking and scratching, in a dose-dependent manner (0.01-10 pmol and 0.01-10 nmol, respectively) in mice. These pronociceptive effects induced by [Tyr(6)] gamma2-MSH(6-12) were inhibited dose-dependently by co-injection of competitive NMDA receptor antagonist D-APV, non-competitive NMDA receptor antagonist MK-801, and nitric oxide (NO) synthase inhibitor L-NAME. However, the tachykinin NK(1) receptor antagonist L-703,606, and tachykinin NK(2) receptor antagonist MEN-10,376, had no influence on pronociceptive effects elicited by [Tyr(6)] gamma2-MSH(6-12). In other groups, [Tyr(6)] gamma2-MSH(6-12)-induced nociceptive responses were bidirectionally regulated by the co-injection of N/OFQ. N/OFQ inhibited nociceptive responses at high doses (0.01-1 nmol), but potentiated the behaviors at low doses (1 fmol-3 pmol). Furthermore, both hyperalgesia and nociceptive responses were enhanced after the co-administration with NOP receptor antagonist [Nphe(1)]N/OFQ(1-13)-NH(2). These results suggest that intrathecal [Tyr(6)] gamma2-MSH(6-12)-induced pronociceptive effects may be mediated through NMDA receptor-NO system in the spinal cord, and demonstrate the interaction between MrgC and N/OFQ-NOP receptor system in pain transmission.


Nociceptors/metabolism , Opioid Peptides/metabolism , Pain/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/metabolism , gamma-MSH/pharmacology , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Hormones/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Spinal , Male , Mice , Narcotic Antagonists , Neurokinin A/analogs & derivatives , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Nociceptors/drug effects , Opioid Peptides/pharmacology , Pain/chemically induced , Pain/physiopathology , Pain Measurement/drug effects , Peptide Fragments , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Opioid/metabolism , Spinal Cord/drug effects , Spinal Cord/physiopathology , Nociceptin Receptor , Nociceptin
13.
Regul Pept ; 156(1-3): 90-5, 2009 Aug 07.
Article En | MEDLINE | ID: mdl-19345242

Neuropeptide S (NPS), a recently identified bioactive peptide through reverse pharmacology approach, was reported to regulate arousal, anxiety, locomotor activity, feeding behaviors and drug reward. NPS receptor (NPSR) mRNA was found in the area related to the descending control system of pain, such as the periaqueductal gray (PAG), raphe nuclei, and lateral parabrachial nucleus (PBN), suggesting a possible role of the NPS-NPSR system in the regulation of pain transmission. In the present study, we evaluated the effects of NPS in pain modulation at the supraspinal level for the first time, using the tail withdrawal test and hot-plate test in mice. NPS (mouse, 0.01-1 nmol) injected intracerebroventricularly (i.c.v.) caused a significant increase of tail withdrawal latency and paw-licking/jumping latency in the tail withdrawal test and the hot-plate test, respectively. Antinociceptive effect elicited by NPS (0.1 nmol, i.c.v.) was not affected by naloxone (i.c.v., 10 nmol co-injection or i.p., 10 mg/kg, 10 min prior to NPS) in both tail withdrawal test and hot-plate test. However, at the doses, naloxone significantly inhibited the antinociceptive effect induced by morphine (i.c.v., 3 nmol). NPS (0.1 nmol, i.c.v.)-induced antinociception was inhibited by co-injection with 10 nmol, but not 3 nmol [D-Cys(tBu)(5)]NPS, a peptidergic antagonist identified more recently, while [D-Cys(tBu)(5)]NPS (3 and 10 nmol) alone induced neither hyperalgesia nor antinociception. These results revealed that NPS could produce antinociception through NPS receptor, but not opioid receptor, and NPS-NPSR system could be a potential target for developing new analgesic drugs.


Analgesics/pharmacology , Neuropeptides/pharmacology , Animals , Mice , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain/drug therapy , Pain/prevention & control
14.
Peptides ; 30(2): 234-40, 2009 Feb.
Article En | MEDLINE | ID: mdl-18992779

Neuropeptide S (NPS), a recently identified bioactive peptide, was reported to regulate arousal, anxiety, motoring and feeding behaviors. NPS precursor and NPS receptor mRNA were found in the amygdala, the ventral tegmental area (VTA) and the substantia nigra, the area thought to modulate rewarding properties of drugs. In the present study, we examined the influence of NPS on the rewarding action of morphine, using the unbiased conditioned place preference (CPP) paradigm. Morphine (1, 3 and 6 nmol, i.c.v.) induced a significant place preference. For testing the effect of NPS on the acquisition of morphine CPP, mice were given the combination of NPS and morphine on the conditioning days, and without drug treatment on the followed test day. To study the effect of NPS on the expression of morphine CPP, mice received the treatment of saline/morphine on the conditioning days, and NPS on the test day, 15 min before the placement in the CPP apparatus. Our results showed that NPS (0.3-10 nmol) alone neither induced place preference nor aversion, however, NPS (1 and 3 nmol) blocked the acquisition of CPP induced by 3 nmol morphine, and acquisition of 6 nmol morphine-induced CPP was also reduced by NPS (6 and 10 nmol). Moreover, the expression of CPP induced by 6 nmol morphine was also inhibited by NPS (0.1, 1 and 10 nmol). These results revealed the involvement of NPS in rewarding activities of morphine, and demonstrated the interaction between NPS system and opioid system for the first time.


Conditioning, Psychological/drug effects , Morphine/pharmacology , Neuropeptides/pharmacology , Animals , Behavior, Animal , Male , Maze Learning/drug effects , Mice , Mice, Inbred Strains , Neuropeptides/administration & dosage
15.
Pharmazie ; 63(9): 676-85, 2008 Sep.
Article En | MEDLINE | ID: mdl-18819522

OBJECTIVE: To investigate the effect and mechanism of nociceptin/orphanin FQ (OFQ) reversing multi-drug resistance of K562/ADM cells in vitro. METHODS: MTT assay, Wright staining, flow cytometry, transmission electron microscope and gel electrophoresis were used to evaluate the effect and mechanism of OFQ in reversing multi-drug resistance of K562/ADM cells. RESULTS: OFQ could time-dependently reverse the ADM resistance of K562/ADM cell. After treatment with OFQ (1 x 10(-7) mol x L(-1)), K562/ADM cells were cultured for 24, 48 and 72 h. The reversal index (RI) was 1.33, 1.42 and 1.53, respectively. Furthermore, OFQ significantly increased the intracellular accumulation of ADM in K562/ADM cells and percentage apoptosis in K562/ADM cells. OFQ down-regulated the level of P-gp time-dependently, while the level of Fas and FasL were up-regulated. There were evidently significant differences compared with the control (P < 0.01). After treating K562/ADM cells with OFQ (1 x 10(-7) mol x L(-1)) and ADM (20 microg x ml(-1)) for 48 hours, the cells showed apoptotic nuclear fragmentation, which was characterized by the appearance of a DNA ladder pattern in genomic DNA gel electrophoresis. CONCLUSION: OFQ can reverse the ADM resistance of K562/ADM cells. The mechanism involves OFQ up-regulating the expression of Fas/FasL, down-regulating the level of P-gp, and decreasing the intracellular level of calcium in K562/ADM cells.


Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Opioid Peptides/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Annexins/metabolism , Calcium/metabolism , Comet Assay , DNA/genetics , DNA Fragmentation/drug effects , Doxorubicin/metabolism , Fas Ligand Protein/metabolism , Flow Cytometry , Fluorescein-5-isothiocyanate , Humans , K562 Cells , Spectrometry, Fluorescence , fas Receptor/metabolism , Nociceptin
16.
Regul Pept ; 142(1-2): 16-23, 2007 Jul 05.
Article En | MEDLINE | ID: mdl-17331598

Cumulative evidence indicates that bone marrow mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating to osteogenic and adipogenic lineages when stimulated under appropriate conditions. Whether OGP(10-14) directly regulates the progenitor cells differentiating into osteoblasts or adipocytes remains unknown. In the present study, we investigated the roles of OGP(10-14) in differentiation along these separate lineages using rat bone marrow MSCs. Our results showed that OGP(10-14) promoted osteogenic differentiation of the stem cells and concurrently inhibited adipocyte formation. OGP(10-14) increased alkaline phosphatase (ALP) activity and mineralized nodule formation, and stimulated osteoblast-specific mRNA expression of core-binding factor 1 (cbfa1). In contrast, OGP(10-14) decreased adipocyte numbers and inhibited adipocyte-specific mRNA expression of peroxisome proliferator-activated receptor-gamma 2 (PPARgamma2). These observations suggest that commitment of MSCs into osteogenic or adipogenic lineages is regulated by OGP(10-14).


Adipocytes/drug effects , Cell Differentiation/drug effects , Endorphins/pharmacology , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/drug effects , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Female , Gene Expression/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Microscopy, Phase-Contrast , NIH 3T3 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
17.
Peptides ; 27(9): 2207-13, 2006 Sep.
Article En | MEDLINE | ID: mdl-16762456

BIBP3226 {(R)-N2-(diphenylacetyl)-N-[(4-hydroxyphenyl)-methyl]-argininamide} was recently shown to display relatively high affinities for neuropeptide FF (NPFF) receptors and exhibit antagonist activities towards NPFF receptors in vitro. The present study was undertaken to investigate the antagonistic effects of BIBP3226 on several in vivo pharmacologic profiles induced by exogenous NPFF and NPVF. (1) BIBP3226 (5 nmol) injected into the third ventricle completely antagonized the hypothermic effects of NPFF (30 nmol) and NPVF (30 nmol) after cerebral administration in mice; (2) BIBP3226 (5 nmol, i.c.v.) prevented the anti-morphine actions of NPFF (10 nmol, i.c.v.) in the mouse tail-flick assay; (3) in urethane-anaesthetized rats, both NPFF (200 nmol/kg, i.v.) and NPVF (200 nmol/kg, i.v.) increased the mean arterial blood pressure, which were significantly reduced by pretreatment with BIBP3226 (500 nmol/kg, i.v.). Collectively, these data suggest that BIBP3226, a mixed antagonist of NPY Y1 and NPFF receptors, shows in vivo antagonistic effects on NPFF receptors. In addition, it seems to be clear that the in vivo pharmacological profiles of NPFF are mediated directly by NPFF receptors.


Arginine/analogs & derivatives , Oligopeptides/antagonists & inhibitors , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Anti-Anxiety Agents/metabolism , Anti-Anxiety Agents/pharmacology , Arginine/pharmacology , Hypothermia/chemically induced , Hypothermia/metabolism , Male , Mice , Oligopeptides/metabolism , Rats , Rats, Wistar , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide Y/metabolism , Tachycardia/metabolism , Time Factors
18.
Regul Pept ; 134(2-3): 75-81, 2006 May 15.
Article En | MEDLINE | ID: mdl-16516988

Two novel ligands for the nociceptin/orphanin FQ (N/OFQ) receptor (NOP), [(pF)Phe4,Aib7, Aib11,Arg14,Lys15]N/OFQ-NH2 (peptide-1) and [Nphe1,(pF)Phe4,Aib7,Aib11,Arg14,Lys15]N/OFQ-NH2 (peptide-2), have been generated by combining different modifications of N/OFQ sequence. In the present study, we investigated the actions of two analogues and compared them with those of N/OFQ in four assays. Peptide-1 mimicked N/OFQ effects in mouse vas deferens and mouse colon and showed similar maximal effects but higher potency relative to N/OFQ. The effects of peptide-1 were sensitive to NOP receptor selective antagonist ([Nphe1]N/OFQ(1-13)-NH2) but not to naloxone in vitro. Peptide-1 (25 pmol, i.c.v.) mimicked the pronociceptive action of N/OFQ (2.5 nmol, i.c.v.) in mouse tail withdrawal assay, displaying higher potency and longer lasting effects. In anesthetized rats, peptide-1 (1 nmol/kg, i.v.) produced a marked decrease in mean arterial pressure, which was comparable to that evoked by i.v. N/OFQ (100 nmol/kg). Peptide-2 did not produce any effect per se but antagonized N/OFQ actions in mouse vas deferens and mouse colon assays. Peptide-2 is active in vivo where it prevented the pronociceptive effect induced by 2.5 nmol N/OFQ i.c.v. in the mouse tail withdrawal assay. Furthermore, peptide-2 at 5 nmol produced alone a robust and long lasting antinociceptive effect. Moreover, peptide-2 (10 and 40 nmol/kg i.v.) didn't produce any effect per se but antagonized hypotensive actions produced by i.v. administration of N/OFQ. Collectively, these findings demonstrate that [(pF)Phe4,Aib7,Aib11, Arg14,Lys15]N/OFQ-NH2 behaves as a highly potent NOP receptor agonist which produces long lasting effects in vivo and [Nphe1,(pF)Phe4,Aib7,Aib11,Arg14,Lys15]N/OFQ-NH2 acts as a pure and competitive antagonist of the NOP receptor.


Opioid Peptides/pharmacology , Receptors, Opioid/drug effects , Animals , Blood Pressure/drug effects , Colon/drug effects , Colon/physiology , Electrophysiology , Female , Male , Mice , Muscle Contraction/drug effects , Naloxone/pharmacology , Narcotic Antagonists , Rats , Rats, Wistar , Receptors, Opioid/agonists , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin Receptor
19.
Brain Res ; 1085(1): 43-8, 2006 Apr 26.
Article En | MEDLINE | ID: mdl-16566906

The pain modulatory properties of melatonin (MT) are generally recognized but the detail of the interaction between melatonin and opioid system in pain regulation is not fully understood. The present study was undertaken to investigate the modulatory effect of melatonin (MT) on the hyperalgesic effect of Orphanin FQ/Nociceptin (OFQ/NC, NC), a member of opioid peptide family. Intracerebroventricular (i.c.v.) administration of NC (10 microg/mouse) induced significant hyperalgesic effect in tail-flick test in mice; i.c.v. (5, 10, 50 microg/mouse) or intraperitoneal (i.p.) (5, 10, 50 mg/kg) co-injection of melatonin dose-dependently reversed NC-induced hyperalgesia and showed a profound analgesic effect. The antihyperalgesia effect of MT could be significantly antagonized by i.c.v. co-injection of luzindole (10 microg/mouse) (an antagonist of MT receptor) or naloxone (10 microg/mouse) (antagonist of traditional opioid receptor). Taken together, all the results suggested that MT could produce a luzindole and naloxone sensitive reversing effect on NC-induced hyperalgesia at supraspinal and peripheral level in mice. The augmentation effect of MT on the traditional opioid system may be one of the mechanisms of this antihyperalgesia action induced by MT. The present work will help to elucidate the mechanism of the pain modulation effect of MT, and also will help to represent new interesting modulating therapeutic targets for the relief of pain.


Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Melatonin/administration & dosage , Opioid Peptides/administration & dosage , Analysis of Variance , Animals , Behavior, Animal , Dose-Response Relationship, Drug , Drug Interactions , Injections, Intraventricular/methods , Injections, Subcutaneous/methods , Male , Mice , Pain Measurement/methods , Nociceptin
20.
Peptides ; 27(6): 1297-304, 2006 Jun.
Article En | MEDLINE | ID: mdl-16337714

To further evaluate the importance of C-terminal modification of neuropeptide FF (NPFF), in the present work, four dansylated NPFF analogues, including two putative agonists (dansyl-PQRFamide and dansyl-GSRFamide) and two putative antagonists (dansyl-PQRamide and dansyl-GSRamide), were synthesized and investigated to address their potencies and efficacies in a series of in vitro and in vivo assays. (1) In the isolated mouse colon bioassay, the four dansylated compounds showed agonistic profiles: both dansyl-GSRFamide (1-10 microM) and dansyl-GSRamide (1-10 microM) dose-dependently caused colonic contractions, which were attenuated by pretreatment with BIBP3226; dansyl-PQRFamide and dansyl-PQRamide evoked modest colonic contractions at a high dose of 50 microM. (2) In urethane-anaesthetized rats, both dansyl-PQRFamide (50-300 nmol/kg, i.v.) and dansyl-GSRFamide (15-50 nmol/kg, i.v.) dose-dependently increased the mean arterial pressure and heart rate in a manner similar to NPFF (50-300 nmol/kg, i.v.); on the contrary, the two putative antagonists (100-800 nmol/kg, i.v.) decreased blood pressure in a dose-dependent manner. All the results suggest that dansyl-PQRFamide and dansyl-GSRFamide are NPFF full agonists; in contrast, dansyl-GSRamide and dansyl-PQRamide behave as agonists in vitro and antagonists in vivo on NPFF receptors. The findings reveal that the C-terminal Phe might be a crucial residue to determine the efficacy. In addition, the novel analogue dansyl-GSRFamide may be developed as a highly potent agonist to investigate the NPFF system.


Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/chemistry , Animals , Biological Assay , Cardiovascular System/drug effects , Colon/drug effects , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Mice , Pressure , Protein Structure, Tertiary , Rats , Rats, Wistar , Spinal Cord/metabolism
...