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










Publication year range
1.
J Pharmacol Sci ; 145(1): 52-59, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33357779

ABSTRACT

DA-9801, a plant-based drug used for the treatment of diabetic neuropathy, is known to improve angiotensin II (Ang II)-induced vascular endothelial cell dysfunction. However, the underlying mechanism is not fully understood. We aimed to determine whether the protective effect of DA-9801 against Ang II-induced endothelial cell dysfunction was mediated via inhibition of endothelial cell inflammation and apoptosis. Ang II-induced oxidative stress was attenuated by pretreatment of human dermal microvascular endothelial cells (HDMECs) with DA-9801. This prevented the Ang II-induced upregulation of NAD(P)H oxidase (the NOX4 and p22phox subunits) and reactive oxygen species. Further, pretreatment of HDMECs with DA-9801 ameliorated Ang II-mediated nuclear factor kappa B activity via prevention of the upregulation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase. It also decreased the Ang II-stimulated increase in inducible nitric oxide synthase (NOS) and decreased endothelial NOS protein expression. DA-9801 decreased Ang II-induced upregulation of intercellular adhesion molecule 1, vascular adhesion molecule, and E-selectin in HDMECs. Moreover, TUNEL and annexin V-FITC fluorescence staining for apoptosis and the activities of caspases 9, 7, and 3 decreased in HDMECs pretreated with DA-9801, indicating that the drug enhanced anti-apoptotic pathways. Thus, DA-9801 modulated Ang II-induced endothelial cell dysfunction via inflammatory and apoptotic pathways.


Subject(s)
Angiotensin II/adverse effects , Apoptosis/drug effects , Endothelial Cells/pathology , Endothelial Cells/physiology , Inflammation/metabolism , Plant Preparations/pharmacology , Cells, Cultured , Dermis/cytology , Humans , Intercellular Adhesion Molecule-1/metabolism , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Alzheimers Res Ther ; 10(1): 11, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29378621

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by deposition of amyloid plaques and disruption of neural circuitry, leading to cognitive decline. Animal models of AD deposit senile plaques and exhibit structural and functional deficits in neurons and neural networks. An effective treatment would prevent or restore these deficits, including calcium dyshomeostasis observed with in-vivo imaging. METHODS: We examined the effects of DA-9803, a multimodal botanical drug, in 5XFAD and APP/PS1 transgenic mice which underwent daily oral treatment with 30 or 100 mg/kg DA-9803 or vehicle alone. Behavioral testing and longitudinal imaging of amyloid deposits and intracellular calcium in neurons with multiphoton microscopy was performed. RESULTS: Chronic administration of DA-9803 restored behavioral deficits in 5XFAD mice and reduced amyloid-ß levels. DA-9803 also prevented progressive amyloid plaque deposition in APP/PS1 mice. Elevated calcium, detected in a subset of neurons before the treatment, was restored and served as a functional indicator of treatment efficacy in addition to the behavioral readout. In contrast, mice treated with vehicle alone continued to progressively accumulate amyloid plaques and calcium overload. CONCLUSIONS: In summary, treatment with DA-9803 prevented structural and functional outcome measures in mouse models of AD. Thus, DA-9803 shows promise as a novel therapeutic approach for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Learning/drug effects , Longitudinal Studies , Male , Mice, Transgenic , Phytotherapy , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Random Allocation
3.
Arch Pharm Res ; 40(2): 268-281, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27885461

ABSTRACT

Dipeptidyl peptidase 4 (DPP4) is an adipokine that interrupts insulin signaling. The resulting insulin resistance exacerbates hepatic steatosis. We previously reported that the novel DPP4 inhibitor evogliptin improves insulin resistance. This study aimed to verify the therapeutic potential of evogliptin for fatty liver. Evogliptin treatment was initiated simultaneously with a high-fat diet (HFD) feeding in normal mice and in a post-24 week HFD-fed rats. In a prevention study, insulin sensitivity was preserved in evogliptin-treated mice after a 16-week treatment. Overall plasma lipid levels stayed lower and hepatic lipid accumulation was drastically suppressed by evogliptin treatment. Evogliptin reduced hepatic expression of Srebf1, a key transcriptional factor for lipogenesis. Additionally, DPP4 inhibitor-treated mice showed less weight gain. In a treatment study, after evogliptin treatment for 14 weeks in pre-established HFD-fed obese rats, weight loss was marginal, while hepatic lipid accumulation and liver damage assessed by measuring plasma aminotransferase levels were completely resolved, suggesting weight loss-independent beneficial effects on fatty liver. Moreover, reduction in plasma non-esterified fatty acids supported the improvement of insulin resistance by evogliptin treatment. Conclusively, our findings suggest that evogliptin treatment ameliorates fatty liver by increasing insulin sensitivity and suppressing lipogenesis.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Fatty Liver/drug therapy , Insulin Resistance , Lipogenesis/drug effects , Piperazines/therapeutic use , Weight Gain/drug effects , Alanine Transaminase/blood , Animals , Blood Glucose , Diet, High-Fat/adverse effects , Dipeptidyl Peptidase 4/metabolism , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/prevention & control , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Triglycerides/blood
4.
BMC Res Notes ; 7: 332, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24890391

ABSTRACT

BACKGROUND: Among the variety of methods used to evaluate locomotor function following a spinal cord injury (SCI), the Basso Mouse Scale score (BMS) has been widely used for mice. However, the BMS mainly focuses on hindlimb movement rather than on graded changes in body support ability. In addition, some of the scoring methods include double or triple criteria within a single score, which likely leads to an increase in the deviation within the data. Therefore we aimed to establish a new scoring method reliable and easy to perform in mice with SCI. FINDINGS: Our Toyama Mouse Score (TMS) was established by rearranging and simplifying the BMS score and combining it with the Body Support Scale score (BSS). The TMS reflects changes in both body support ability and hindlimb movement. The definition of single score is made by combing multiple criteria in the BMS. The ambiguity was improved in the TMS. Using contusive SCI mice, hindlimb function was measured using the TMS, BMS and BSS systems. The TMS could distinguish changes in hindlimb movements that were evaluated as the same score by the BMS. An analysis of the coefficient of variation (CV) of score points recorded for 11 days revealed that the CV for the TMS was significantly lower than the CV obtained using the BMS. A variation in intra evaluators was lower in the TMS than in the BMS. CONCLUSION: These results suggest that the TMS may be useful as a new reliable method for scoring locomotor function for SCI models.


Subject(s)
Locomotion , Spinal Cord Injuries/physiopathology , Animals , Female , Hindlimb/physiopathology , Male , Mice
5.
Life Sci ; 90(1-2): 21-9, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22056373

ABSTRACT

AIM: To characterize the pharmacodynamic profile of DA-1229, a novel dipeptidyl peptidase (DPP) 4 inhibitor. MAIN METHODS: Enzyme inhibition assays against DPP4, DPP8 and DPP9. Antidiabetic effects of DA-1229 in HF-DIO mice and young db/db mice. KEY FINDINGS: DA-1229 was shown to potently inhibit the DPP4 enzyme in human and murine soluble forms and the human membrane-bound form with IC(50) values of 0.98, 3.59 and 1.26 nM, respectively. As a reversible and competitive inhibitor, DA-1229 was more selective to human DPP4 (6000-fold) than to human DPP8 and DPP9. DA-1229 (0.1-3mg/kg) dose-dependently inhibited plasma DPP4 activity, leading to increased levels of plasma GLP-1 and insulin, and thereby lowering blood glucose levels in mice. In high fat diet-fed (HF) mice, a single oral dose of 100mg/kg of DA-1229 reduced plasma DPP4 activity by over 80% during a 24h period. Long-term treatment with DA-1229 for 8 weeks revealed significant improvements in glucose intolerance and insulin resistance, accompanied by significant body weight reduction. However, it remains unclear whether there is a direct causal relationship between DPP4 inhibition and body weight reduction. In young db/db mice, the DA-1229 treatment significantly reduced blood glucose excursions for the first 2 weeks, resulting in significantly lower levels of HbA1c at the end of the study. Furthermore, the pancreatic insulin content of the treatment group was significantly higher than that of the db/db control. SIGNIFICANCE: DA-1229 as a novel and selective DPP4 inhibitor improves the insulin sensitivity in HF mice and delays the onset of diabetes in young db/db mice.


Subject(s)
Diabetes Mellitus/drug therapy , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin Resistance/physiology , Piperazines/therapeutic use , Animals , CHO Cells , Cricetinae , Cricetulus , Diabetes Mellitus/blood , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperazines/chemistry , Random Allocation , Time Factors
6.
Bioorg Med Chem Lett ; 21(12): 3809-12, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21570283

ABSTRACT

A series of ß-amino amide containing substituted piperazine-2-one derivatives was synthesized and evaluated as inhibitors of dipeptidyl pepdidase-4 (DPP-4) for the treatment of type 2 diabetes. As results of intensive SAR study of the series, (R)-4-[(R)-3-amino-4-(2,4,5-trifluorophenyl)-butanoyl]-3-(t-butoxymethyl)-piperazin-2-one (DA-1229) displayed potent DPP-4 inhibition pattern in several animal models, was selected for clinical development.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Piperazines/chemical synthesis , Administration, Oral , Animals , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Piperazines/chemistry , Piperazines/pharmacology , Rats , Structure-Activity Relationship
7.
Eur J Pharmacol ; 650(2-3): 673-81, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20974124

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) γ is known to be a key regulator of insulin resistance. PAM-1616 is a novel, non-thiazolidinedione small molecule compound synthesized in Dong-A Research Center. In this study, we characterized the pharmacological and safety profiles of PAM-1616 as a selective PPARγ modulator. PAM-1616 selectively binds to human PPARγ (IC(50), 24.1±5.6 nM) and is a partial agonist for human PPARγ with an EC(50) of 83.6±43.7 nM and a maximal response of 24.9±7.1% relative to the full agonist, rosiglitazone. PAM-1616 was selective for human PPARγ than for human PPARα (EC(50), 2658±828 nM) without activating human PPARδ, which makes it a selective modulator of PPARγ. Treatment of high fat diet-induced obese C57BL/6J mice with PAM-1616 for 21 days improved HOMA-IR. Furthermore, PAM-1616 significantly improved hyperglycemia in db/db mice with little side effect when orally administered at a dose of 1 mg/kg/day for 28 days. Intriguingly, PAM-1616 was seen to increase the gene expression of inducible glucose transporter (GLUT4), while it partially induced that of a fatty acid carrier, aP2 in 3T3-L1 adipocytes, and it also showed partial recruitment of an adipogenic cofactor, TRAP220 as compared to rosiglitazone. PAM-1616 did not cause a significant increase in plasma volume of ICR mice when orally administered at a dose of 10 mg/kg/day for 9 days. PAM-1616 increased the expression of fluid retention-inducing genes such as serum/glucocorticoid-regulated kinase (SGK)-1 to a lesser extent as compared to rosiglitazone in human renal epithelial cells. These results suggest that PAM-1616 acts as a selective modulator of PPARγ with excellent antihyperglycemic property. The differential modulation of target gene by PAM-1616 might contribute to the improved side effect profiles.


Subject(s)
Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , PPAR gamma/agonists , Phenylpropionates/therapeutic use , Thiophenes/therapeutic use , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cells, Cultured , Humans , Hyperglycemia/metabolism , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR delta/genetics , PPAR delta/metabolism , Phenylpropionates/adverse effects , Phenylpropionates/pharmacology , Rosiglitazone , Thiazolidinediones/pharmacology , Thiophenes/adverse effects , Thiophenes/pharmacology , Water-Electrolyte Balance/drug effects
8.
Diabetes Res Clin Pract ; 91(1): 72-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21093089

ABSTRACT

We studied the effect of a novel dipeptidyl peptidase IV (DPP IV) inhibitor, DA-1229, on blood glucose profile and pancreatic ß-cell mass in established diabetes after streptozotocin (STZ) treatment. Mice that developed diabetes after administration of STZ 100mg/kg were treated with DA-1229 for 13 weeks. DA-1229 significantly reduced plasma DPP IV activity, and enhanced glucagon-like peptide 1 (GLP-1) levels. In STZ-treated mice fed DA-1229 (STZ-DA), blood glucose levels were significantly lower than those in diabetic mice fed normal chow (STZ-NC). Basal and glucose-stimulated insulin secretion and glucose tolerance assessed by intraperitoneal glucose tolerance test were significantly improved by DA-1229 administration. Volume density of ß-cell was significantly increased in STZ-DA mice compared to STZ-NC mice, suggesting that DA-1229-mediated amelioration of established diabetes was due to beneficial effect of DA-1229 on ß-cell mass. The number of replicating ß-cells and that of scattered small ß-cell unit representing ß-cell neogenesis were significantly increased in STZ-DA mice compared to STZ-NC mice, explaining increased ß-cell mass by DA-1229. The expression of PDX-1, a downstream mediator of GLP-1 action, was increased in islets of STZ-DA mice compared to STZ-NC mice. These results suggest a therapeutic potential of DA-1229 in diabetes, particularly that associated with decreased ß-cell mass.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Insulin-Secreting Cells/drug effects , Piperazines/pharmacology , Piperazines/therapeutic use , Animals , Blood Glucose/analysis , Cell Count , Cell Size/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Dipeptidyl Peptidase 4/blood , Glucagon-Like Peptide 1/blood , Glucose Intolerance/drug therapy , Homeodomain Proteins/metabolism , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Regeneration/drug effects , Streptozocin/toxicity , Trans-Activators/metabolism
9.
Arch Pharm Res ; 32(5): 721-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19471887

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) gamma is known to be a key regulator of insulin resistance. PAR-1622 is a novel small molecule compound synthesized in Dong-A research center. In this study, we characterized the pharmacological profiles of PAR-1622, a selective partial activator of PPARgamma. In transient transactivation assays, PAR-1622 [(S)-2-ethoxy-3(4-(5-(4-(5-(methoxymethyl)isoxazol-3-yl)phenyl)-3-methylthiophen-2-yl)methoxy)phenyl)propanoic acid] showed a partial activator against human PPARgamma with an EC(50) of 41 nM and a maximal response of 37% relative to the full agonist rosiglitazone without activating human PPARdelta. PAR-1622 was 56 folds more selective for human PPARgamma than for human PPARalpha (EC(50), 2304 nM), which means that it is a selective partial activator of PPARgamma. PAR-1622 also showed a partial activator against mouse PPARgamma with an EC(50) of 427 nM and a maximal response was 57% of that of rosiglitazone. INT-131, a selective PPARgamma partial agonist in clinical stage, also was a partial activator against human PPARgamma with an EC(50) of 83 nM and a maximal response achieved by INT-131 was 49% of that observed with full agonist rosiglitazone. In functional assays using human mesenchymal stem cells, PAR-1622 induced adipocyte differentiation, which was 3-fold more potent with a comparable maximum response compared to INT-131. Furthermore, PAR-1622 significantly improved hyperglycemia in db/db when orally administered at a dose of 1 mg/kg/day for 5 days. In hemodilution assays with Evans Blue, rosiglitazone significantly increased the plasma volume in ICR mice that were orally administered 30 mg/kg/day for 9 days; however, PAR-1622 showed no significant effects on plasma volume, similar to INT-131. These results suggest that PAR-1622 is a selective partial activator of PPARgamma and has excellent antihyperglycemic activities and a broad safety profile for fluid retention.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Isoxazoles/pharmacology , PPAR gamma/agonists , Propionates/pharmacology , Thiophenes/pharmacology , Water-Electrolyte Balance/drug effects , Adipogenesis/drug effects , Administration, Oral , Animals , Blood Glucose/drug effects , Blood Volume/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Partial Agonism , Genes, Reporter , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/toxicity , Isoxazoles/administration & dosage , Isoxazoles/toxicity , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred ICR , PPAR gamma/genetics , PPAR gamma/metabolism , Propionates/administration & dosage , Propionates/toxicity , Rosiglitazone , Thiazolidinediones/pharmacology , Thiophenes/administration & dosage , Thiophenes/toxicity , Transfection
10.
Eur J Pharmacol ; 589(1-3): 58-65, 2008 Jul 28.
Article in English | MEDLINE | ID: mdl-18550051

ABSTRACT

MK801 (dizocilpine) induces selective neurotoxic effects in the retrosplenial cortex, ranging from neuronal vacuolization to irreversible neurodegeneration depending on the dose administered. Although lamotrigine prevents MK801-induced neuronal vacuolization in the retrosplenial cortex 4 h after injection, it is not clear whether lamotrigine attenuates the subsequent neurodegeneration that occurs 3-4 days later. Because early growth response factor-1 (egr-1) plays a key role in neurodegeneration and its expression is induced in the retrosplenial cortex following MK801 treatment, it is possible that lamotrigine may attenuate MK801-induced neurodegeneration via inhibition of egr-1 expression in the retrosplenial cortex. To address this issue, we treated rats with lamotrigine (10 or 20 mg/kg) followed by MK801 (2 mg/kg) and measured changes in the levels of egr-1 mRNA and immunoreactivity in the retrosplenial cortex and other brain regions 3 h later. We also evaluated the effects of these treatments on neurodegeneration 4 days following treatment using Fluoro-Jade B staining. MK801 treatment increased egr-1 mRNA and immunoreactivity in the restrosplenial, cingulate, entorhinal and piriform cortices, but decreased levels in hippocampal subfields. These MK801-induced changes in egr-1 expression were significantly inhibited by lamotrigine pretreatment. In addition, MK801-induced neurodegeneration in the retrosplenial cortex was partially blocked by lamotrigine pretreatment in a dose dependent manner. These results demonstrate that lamotrigine pretreatment prevents the MK801-induced upregulation of egr-1 expression in a region-selective manner, and suggest that this effect may contribute, in part, to the attenuation of MK801-induced neurodegeneration in the retrosplenial cortex.


Subject(s)
Cerebral Cortex/drug effects , Early Growth Response Protein 1/metabolism , Hippocampus/drug effects , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , RNA, Messenger/metabolism , Triazines/pharmacology , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Dizocilpine Maleate , Dose-Response Relationship, Drug , Early Growth Response Protein 1/genetics , Hippocampus/metabolism , Immunohistochemistry , Lamotrigine , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
Korean J Physiol Pharmacol ; 12(3): 117-23, 2008 Jun.
Article in English | MEDLINE | ID: mdl-20157404

ABSTRACT

Although growth associated protein-43 (GAP-43) is known to play a significant role in the regulation of axonal growth and the formation of new neuronal connections in the hippocampus, there is only a few studies on the effects of acute stress on GAP-43 mRNA expression in the hippocampus. Moreover, the effects of repeated citalopram treatment on chronic mild stress (CMS)-induced changes in GAP-43 mRNA expression in the hippocampus have not been explored before. To explore this question, male rats were exposed to acute immobilization stress or CMS. Also, citalopram was given prior to stress everyday during CMS procedures. Acute immobilization stress significantly increased GAP-43 mRNA expression in all subfields of the hippocampus, while CMS significantly decreased GAP-43 mRNA expression in the dentate granule cell layer (GCL). Repeated citalopram treatment decreased GAP-43 mRNA expression in the GCL compared with unstressed controls, but this decrease was not further potentiated by CMS exposure. Similar decreases in GAP-43 mRNA expression were observed in CA1, CA3 and CA4 areas of the hippocampus only after repeated citalopram treatment in CMS-exposed rats. This result indicates that GAP-43 mRNA expression in the hippocampus may differently respond to acute and chronic stress, and that repeated citalopram treatment does not change CMS-induced decreases in GAP-43 mRNA expression in the GCL.

12.
Neuropharmacology ; 50(7): 824-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16504218

ABSTRACT

Accumulating evidence suggests that dysregulation of corticotropin-releasing factor (CRF) may play a role in depression and that this dysregulation may be corrected by antidepressant drug treatment. Here, we examined whether chronic mild stress (CMS) alters CRF mRNA levels in stress-related brain areas including the bed nucleus of the stria terminalis (BNST) and the central nucleus of amygdala (CeA), and whether repeated tianeptine treatment can attenuate CMS-induced changes in CRF mRNA levels. Male rats were exposed to CMS for 19 days, and control animals were subjected to brief handling. Both groups were injected daily with tianeptine or saline. CMS significantly increased CRF mRNA levels in the dorsal BNST (dBNST), but not in other areas. Repeated tianeptine treatment prevented the CMS-induced increase in CRF mRNA levels in the dBNST, and reduced CRF mRNA levels in dBNST in non-stressed controls. Moreover, repeated tianeptine treatment significantly decreased CRF mRNA levels in the ventral BNST and CeA of non-stressed controls as well as CMS-exposed rats. These results show that CMS induces a rather selective increase of CRF mRNA in the dBNST. In addition, these results suggest that repeated tianeptine treatment diminishes the basal activity of CRF neurons and reduces their sensitivity to stress.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Gene Expression Regulation/drug effects , RNA, Messenger/biosynthesis , Stress, Physiological/metabolism , Thiazepines/administration & dosage , Animals , Corticotropin-Releasing Hormone/genetics , Gene Expression Regulation/physiology , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Stress, Physiological/drug therapy , Stress, Physiological/genetics
13.
Exp Mol Med ; 38(1): 44-54, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16520552

ABSTRACT

New-born cells continue to proliferate and survive to become mature granule cells in adult rat hippocampus. Although this process, known as neurogenesis, is inhibited by acute stress, it is not clear whether chronic stress affects neurogenesis. To determine whether chronic mild stress (CMS) influences neurogenesis in the adult rat hippocampus, male Sprague-Dawley rats were exposed to CMS and administered bromodeoxyuridine (BrdU) before or after CMS to observe the survival/differentiation or proliferation of new-born cells, respectively. In addition, we measured brain-derived neurotrophic factor (BDNF) mRNA in the granule cell layer (GCL) of the hippocampus, because BDNF is known to play an important role in the survival of new-born cells. CMS significantly decreased the survival of new-born cells in the GCL, but did not influence the proliferation or differentiation of new-born cells. CMS did not affect the proliferation and survival of new-born cells in the hilus. In addition, CMS did not change BDNF mRNA levels in the GCL. These results demonstrate that CMS reduces the survival of new-born cells but not of their proliferation, suggesting that repeated mild stress could influence a part of neurogenesis, but not the whole part of neurogenesis. These results raise the possibility that the survival of new-born cells may be suppressed in the presence of normal BDNF mRNA levels in GCL.


Subject(s)
Bromodeoxyuridine/administration & dosage , Hippocampus/pathology , Stress, Physiological/physiopathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calbindins , Cell Proliferation , Cell Survival , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , In Situ Hybridization , Male , Microscopy, Confocal , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical , Rhodamines , S100 Calcium Binding Protein G/metabolism , Stress, Physiological/pathology
14.
Peptides ; 26(11): 2246-51, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16269353

ABSTRACT

Doxapram causes panic anxiety in humans. To determine whether doxapram alters corticotropin-releasing factor (CRF) expression in the central nucleus of the amygdala (CeA), paraventricular nucleus of hypothalamus (PVN), or bed nucleus of the stria terminalis (BNST), we used immunohistochemistry to measure CRF peptide in these brain areas after doxapram injection. Doxapram injection significantly increased CRF-like immunoreactivity (CRF-IR) within the CeA, but not in the BNST or PVN, and this increase was significant 2h after injection. In addition, doxapram significantly increased CRF mRNA expression within the CeA, and this was most prominent 30min after injection. These results suggest that doxapram selectively increases CRF expression within the CeA, and that this is mediated by increased CRF gene transcription. This increase in CRF-IR within the CeA might explain the doxapram-induced anxiety reaction.


Subject(s)
Amygdala/metabolism , Central Nervous System Stimulants/adverse effects , Corticotropin-Releasing Hormone/biosynthesis , Doxapram/adverse effects , Gene Expression Regulation/drug effects , Amygdala/pathology , Animals , Central Nervous System Stimulants/administration & dosage , Doxapram/administration & dosage , Immunohistochemistry , In Situ Hybridization , Male , Panic Disorder/chemically induced , Panic Disorder/metabolism , Panic Disorder/pathology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Septal Nuclei/metabolism , Septal Nuclei/pathology
15.
Mol Cells ; 19(1): 74-80, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15750343

ABSTRACT

Stress is known to inhibit granule cell proliferation in the hippocampus. However, recent studies suggest that the commonly used dose of bromodeoxyuridine (BrdU) is insufficient to label all fractions of granule cells. Furthermore, stress-induced changes in BrdU availability may influence the labeling of newly born cells. To investigate whether changes in BrdU availability affect measurements of stress-induced granule cell proliferation, granule cell proliferation was assessed using injection of high doses of BrdU before and after restraint stress lasting 1 h. In addition, to determine whether stress-induced changes in plasma corticosterone levels were influenced by the BrdU, time-dependent changes in plasma corticosterone levels over 2 h after BrdU injection were compared with total accumulated plasma corticosterone levels [as determined by areas under the curve (AUC)]. Restraint stress significantly reduced the numbers of BrdU-labeled cells and clusters in the granule cell layer (GCL) of rats that received BrdU after stress, and decreases of similar magnitude were observed when the rats were given BrdU before stress. BrdU injection enhanced the stress-induced plasma corticosterone response, but there was no difference between the mean AUCs of plasma corticosterone levels of animals injected with BrdU before or after stress. These observations suggest that restraint stress decreases granule cell proliferation, and that this may be influenced by the extent and duration of plasma corticosterone increases rather than by changes in the availability of BrdU.


Subject(s)
Bromodeoxyuridine/administration & dosage , Hippocampus/pathology , Stress, Physiological/physiopathology , Animals , Cell Proliferation , Corticosterone/blood , Hippocampus/cytology , Hippocampus/growth & development , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical , Stress, Physiological/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...