Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
PLoS One ; 13(2): e0193289, 2018.
Article in English | MEDLINE | ID: mdl-29466448

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disease generally caused by reading frame disrupting mutations in the DMD gene resulting in loss of functional dystrophin protein. The reading frame can be restored by antisense oligonucleotide (AON)-mediated exon skipping, allowing production of internally deleted, but partially functional dystrophin proteins as found in the less severe Becker muscular dystrophy. Due to genetic variation between species, mouse models with mutations in the murine genes are of limited use to test and further optimize human specific AONs in vivo. To address this we have generated the del52hDMD/mdx mouse. This model carries both murine and human DMD genes. However, mouse dystrophin expression is abolished due to a stop mutation in exon 23, while the expression of human dystrophin is abolished due to a deletion of exon 52. The del52hDMD/mdx model, like mdx, shows signs of muscle dystrophy on a histological level and phenotypically mild functional impairment. Local administration of human specific vivo morpholinos induces exon skipping and dystrophin restoration in these mice. Depending on the number of mismatches, occasional skipping of the murine Dmd gene, albeit at low levels, could be observed. Unlike previous models, the del52hDMD/mdx model enables the in vivo analysis of human specific AONs targeting exon 51 or exon 53 on RNA and protein level and muscle quality and function. Therefore, it will be a valuable tool for optimizing human specific AONs and genome editing approaches for DMD.


Subject(s)
Base Sequence , Dystrophin , Exons , Oligodeoxyribonucleotides, Antisense , Sequence Deletion , Animals , Drug Evaluation, Preclinical , Dystrophin/genetics , Dystrophin/metabolism , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology
2.
BMC Complement Altern Med ; 16: 240, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27457480

ABSTRACT

BACKGROUND: Pain, the most common reasons for physician consultation, is a major symptom in many medical conditions that can significantly interfere with a person's life quality and general functioning. Almost all painkillers have its untoward effects. Therefore, seeking for a safe medication for pain relieve is notable nowadays. Paeonia lactiflora is a well-known traditional Chinese medicine. Paeoniflorin is an active component found in Paeonia lactiflora, which has been reported to inhibit formalin-induced nociceptive behavior in mice. Aims of this present study were to investigate effects of paeoniflorin on excitatory amino acid agonist- or high-dose morphine-induced nociceptive behaviors in mice. RESULTS: Paeoniflorin (100, 200, 500 nmol, i.c.v.) alone and combined with glutamatergic antagonists (MK-801 14.8 pmol, or NBQX 5 nmol, i.t.) inhibited nociception. Those agents also inhibited the clonic seizure-like excitation induced by high-dose morphine (250 nmol, i.t) in mice. Antisense oligodeoxynucleotides of NMDA receptor subunits NR1, NR2A, NR2B significantly enhanced the inhibition of paeoniflorin on excitatory amino acid-and high-dose morphine-induced nociception. Docking energy data revealed that paeoniflorin had stronger binding activity in NR2A and NR2B than NR2C of NMDA receptors. CONCLUSIONS: Results of this study indicate that paeoniflorin-induced inhibition of excitatory amino acid agonist- and high-dose morphine-induced nociceptive behaviors might be due to modulation of NMDA receptors, specifically the NR2B subunit.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Excitatory Amino Acid Agonists/pharmacology , Glucosides/pharmacology , Monoterpenes/pharmacology , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Analgesics , Animals , Male , Mice , Mice, Inbred ICR , Oligodeoxyribonucleotides, Antisense/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics
3.
J Nanosci Nanotechnol ; 16(2): 1370-4, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27433588

ABSTRACT

A series of pluronic grafted dendritic alpha,epsilon-poly(L-lysine)s (DPL-PF127) were synthesized by a conjugation reaction and evaluated the potential use of DPL-PF127 as a delivery agent of antisense oligonucleotide into A375 B3 cells. The structural features of the DPL-PF127 were identified by NMR and FT-IR. The number of pluronic F127 on DPL surface, determined by fluorescamine assay, increased proportionally to the mole ratio between DPL and activated PF127 in reaction. DPL- PF127 showed the physical properties of decrease in zetapotential and increase in size as the mole ratio of PF127 to DPL increased. The complex formation of DPL-PF127 with oligonucleotide was confirmed by running capillary zone electrophoresis (CZE) and agarose gel electrophoresis. DPL-PF127, prepared at the mole ratio of 1:10 in reaction, was the most suitable as a delivery adjuvant of oligonucleotide. In addition, DPL-PF127/oligonucleotide complexes were taken into A375B3 cell without cellular toxicity and delivered antisense oligonucleotide into cell.


Subject(s)
Drug Carriers , Oligodeoxyribonucleotides, Antisense , Poloxamer , Polylysine , Cell Line , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Oligodeoxyribonucleotides, Antisense/chemistry , Oligodeoxyribonucleotides, Antisense/pharmacokinetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Poloxamer/chemistry , Poloxamer/classification , Poloxamer/pharmacokinetics , Poloxamer/pharmacology , Polylysine/chemistry , Polylysine/pharmacokinetics , Polylysine/pharmacology
4.
J Neurochem ; 132(2): 194-205, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25270764

ABSTRACT

Ceftriaxone(Cef) selectively increases the expression of glial glutamate transporter-1 (GLT-1), which was thought to be neuroprotective in some circumstances. However, the effect of Cef on glutamate uptake of GLT-1 was mostly assayed using in vitro studies such as primary neuron/astrocyte cultures or brain slices. In addition, the effect of Cef on neurons in different ischemic models was still discrepant. Therefore, this study was undertaken to observe the effect of Cef on neurons in global brain ischemia in rats, and especially to provide direct evidence of the up-regulation of GLT-1 uptake for glutamate contributing to the neuronal protection of Cef against brain ischemia. Neuropathological evaluation indicated that administration of Cef, especially pre-treatment protocols, significantly prevented delayed neuronal death in hippocampal CA1 subregion normally induced by global brain ischemia. Simultaneously, pre-administration of Cef significantly up-regulated the expression of GLT-1. Particularly, GLT-1 uptake assay with (3) H-glutamate in living cells from adult rats showed that up-regulation in glutamate uptake accompanied up-regulated GLT-1 expression. Inhibition of GLT-1 by antisense oligodeoxynucleotides or dihydrokainate significantly inhibited the Cef-induced up-regulation in GLT-1 uptake and the neuroprotective effect against global ischemia. Thus, we may conclude that Cef protects neurons against global brain ischemia via up-regulation of the expression and glutamate uptake of GLT-1. Glutamate uptake by glial glutamate transporter-1 (GLT-1) is the principal way to regulate extracellular glutamate homeostasis in central nervous system. Over-accumulation of glutamate results in excitotoxicity and injures neurons after cerebral ischemia. Ceftriaxone up-regulates GLT-1 expression and uptake of glutamate, diminishes the excitotoxicity of glutamate and then protects neurons against global brain ischemia.


Subject(s)
Brain Ischemia/drug therapy , Ceftriaxone/therapeutic use , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Neuroprotective Agents/therapeutic use , Animals , Biological Transport/drug effects , Brain Ischemia/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Ceftriaxone/administration & dosage , Ceftriaxone/pharmacology , Drug Evaluation, Preclinical , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Knockdown Techniques , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Random Allocation , Rats , Rats, Wistar , Up-Regulation
5.
J Neuroendocrinol ; 24(11): 1412-22, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22681480

ABSTRACT

Galanin-like peptide (GALP) is a known mediator of metabolism and reproduction; however, the role that GALP plays in the onset of puberty is unknown. First, we tested the hypothesis that central GALP administration could rescue puberty in food-restricted weanling rats. GALP treatment in food-restricted rats of both sexes rescued the timing of the onset of puberty to that seen in ad lib. fed controls. Second, we tested whether GALP translation knocked-down in ad lib. fed, prepubertal rats would alter the timing of puberty. Knock-down females, but not males, showed a significant (P < 0.01) delay in the onset of puberty compared to controls. Third, we sought evidence that the role of GALP in pubertal onset is mediated by the kisspeptin system. In situ hybridisation analyses showed a significant (P < 0.01) reduction in Kiss1 mRNA within the hypothalamic arcuate nucleus in food-restricted rats compared to ad lib. fed controls and this reduction was prevented with i.c.v. GALP administration. Furthermore, analyses of Fos-immunoreactivity (-IR) after i.c.v. GALP treatment did not elicit Fos-IR within any kisspeptin neurones, nor are GALP and kisspeptin peptides or mRNA colocalised. These data demonstrate that hypothalamic GALP infusion maintained the onset of puberty in food-restricted weanling rats, although probably not via direct innervation of kisspeptin neurones.


Subject(s)
Caloric Restriction/adverse effects , Galanin-Like Peptide/administration & dosage , Hypothalamus/drug effects , Sexual Maturation/drug effects , Animals , Female , Food , Galanin-Like Peptide/genetics , Galanin-Like Peptide/metabolism , Galanin-Like Peptide/pharmacology , Hypothalamus/metabolism , Infusions, Intraventricular , Kisspeptins/administration & dosage , Kisspeptins/genetics , Kisspeptins/metabolism , Kisspeptins/pharmacology , Male , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Long-Evans , Sexual Maturation/genetics , Sexual Maturation/physiology , Weaning
6.
DNA Cell Biol ; 31(2): 164-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21793718

ABSTRACT

Proto-oncogenes are involved in cell growth, proliferation, and differentiation. In the present study, we investigated the roles and mediating pathways of proto-oncogenes c-erbB(2) and c-myb in mouse oocyte maturation by RT-PCR, real-time quantitative PCR, western blot, and recombinant proto-oncogene protein microinjection. Results showed that both c-erbB(2) and c-myb antisense oligodeoxynucleotides (c-erbB(2) ASODN and c-myb ASODN) inhibited germinal vesicle breakdown and the first polar body extrusion in a dose-dependent manner. However, microinjection of recombinant c-erbB(2) or c-myb protein into germinal vesicle stage oocytes stimulated oocyte meiotic maturation. In addition, the expression of c-erbB(2) and c-myb mRNA was detected in oocytes; and c-erbB(2) ASODN and c-myb ASODN inhibited c-erbB(2) mRNA and c-myb mRNA expression, respectively. Maturation promoting factor (MPF) inhibitor roscovitine did not affect the expression of c-erbB(2) mRNA and c-myb mRNA, but blocked the effects of recombinant c-erbB(2) and c-myb protein-induced oocyte maturation. Further, cyclin B1 protein expression in oocytes was remarkably inhibited by c-erbB(2) ASODN, c-myb ASODN, and roscovitine. Nonsense tat ODN had no effect on the expression of c-erbB(2), c-myb, and cyclin B1. These results suggest that c-erbB(2) and c-myb may induce oocyte maturation through mediating a pathway involving the activation of MPF.


Subject(s)
Genes, erbB-2/physiology , Genes, myb/physiology , Maturation-Promoting Factor/genetics , Oocytes/physiology , Animals , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Enzymologic/drug effects , Genes, erbB-2/drug effects , Genes, erbB-2/genetics , Genes, myb/drug effects , Genes, myb/genetics , Maturation-Promoting Factor/metabolism , Mesothelin , Mice , Microinjections , Oligodeoxyribonucleotides, Antisense/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Oogenesis/drug effects , Oogenesis/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myb/administration & dosage , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Purines/pharmacology , Receptor, ErbB-2/administration & dosage , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Roscovitine , Transcriptional Activation
7.
J Neurosci ; 31(42): 14835-40, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22016516

ABSTRACT

The transcriptional coactivator PGC-1α is a key regulator of cellular energy expenditure in peripheral tissues. Recent studies report that PGC-1α-null mice develop late-onset obesity and that the neuronal inactivation of PGC-1α causes increased food intake. However, the exact role of PGC-1α in the CNS remains unclear. Here we show that PGC-1α directly regulates the expression of the hypothalamic neuropeptide oxytocin, a known central regulator of appetite. We developed a unique genetic approach in the zebrafish, allowing us to monitor and manipulate PGC-1α activity in oxytocinergic neurons. We found that PGC-1α is coexpressed with oxytocin in the zebrafish hypothalamus. Targeted knockdown of the zebrafish PGC-1α gene activity caused a marked decrease in oxytocin mRNA levels and inhibited the expression of a transgenic GFP reporter driven by the oxytocin promoter. The effect of PGC-1α loss of function on oxytocin gene activity was rescued by tissue-specific re-expression of either PGC-1α or oxytocin precursor in zebrafish oxytocinergic neurons. PGC-1α activated the oxytocin promoter in a heterologous cell culture system, and overexpression of PGC-1α induced ectopic expression of oxytocin in muscles and neurons. Finally, PGC-1α forms an in vivo complex with the oxytocin promoter in fed but not fasted animals. These findings demonstrate that PGC-1α is both necessary and sufficient for the production of oxytocin, implicating hypothalamic PGC-1α in the direct activation of a hypothalamic hormone known to control energy intake.


Subject(s)
Heat-Shock Proteins/metabolism , Hypothalamus/cytology , Neurons/metabolism , Oxytocin/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Animals, Genetically Modified , Cell Line, Transformed , Chromatin Immunoprecipitation/methods , Computational Biology , Embryo, Nonmammalian , Fasting/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/genetics , Humans , Hypothalamus/metabolism , Male , Mice , Neurons/drug effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Oxytocin/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger , Transcription Factors/genetics , Transfection/methods , Zebrafish
8.
Behav Brain Res ; 222(1): 230-5, 2011 Sep 12.
Article in English | MEDLINE | ID: mdl-21458499

ABSTRACT

Ongoing epidemiological studies estimate that greater than 60% of the adult US population may be categorized as either overweight or obese. There is a growing appreciation that the complications of obesity extend to the central nervous system (CNS) and may result in increased risk for neurological co-morbidities like depressive illness. One potential mechanistic mediator linking obesity and depressive illness is the adipocyte derived hormone leptin. We previously demonstrated that lentivirus-mediated downregulation of hypothalamic insulin receptors increases body weight, adiposity and plasma leptin levels, which is consistent with features of the metabolic syndrome. Using this novel model of obesity, we examined performance in the forced swim test (FST), the sucrose preference test and the elevated plus maze (EPM), approaches that are often used as measures of depressive-like and anxiety-like behaviors, in rats that received third ventricular injections of either an insulin receptor antisense lentivirus (hypo-IRAS) or a control lentivirus (hypo-Con). Hypo-IRAS rats exhibited significant increases in immobility time and corresponding decreases in active behaviors in the FST and exhibited anhedonia as measured by decreased sucrose intake compared to hypo-Con rats. Hypo-IRAS rats also exhibited increases in anxiety-like behaviors in the EPM. Plasma, hippocampal and amygdalar brain-derived neurotrophic factor (BDNF) levels were reduced in hypo-IRAS rats, suggesting that the obesity/hyperleptinemic phenotype may elicit this behavioral phenotype through modulation of neurotrophic factor expression. Collectively, these data support the hypothesis for an increased risk for mood disorders in obesity, which may be related to decreased expression of hippocampal and amygdalar BDNF.


Subject(s)
Depression/etiology , Down-Regulation/physiology , Hypothalamus/metabolism , Receptor, Insulin/metabolism , Adiposity/drug effects , Adiposity/physiology , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/physiology , Depression/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Food Preferences/drug effects , Food Preferences/physiology , Genetic Vectors/physiology , Hypothalamus/drug effects , Leptin/blood , Male , Maze Learning/drug effects , Maze Learning/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Insulin/biosynthesis , Swimming/psychology , Triglycerides/blood
9.
Biochem Cell Biol ; 88(4): 687-95, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20651841

ABSTRACT

Early growth response 1 (Egr-1) over-expression has been demonstrated in myocardial ischemia-reperfusion injury, which is closely associated with endothelial dysfunction. In the present study we investigated the expression of Egr-1 on cultured cardiac microvascular endothelial cells (CMECs) to help define the mechanism of myocardial ischemia-reperfusion injury. A model of cultured CMECs exposed to hypoxia-reoxygenation was developed in which synthesized Egr-1 sense and antisense oligodeoxyribonucleotide were transfected into the cells. The expression of Egr-1 was examined by Western blot analysis. Lactate dehydrogenase, malondialdehyde, superoxide dismutase, tumor necrosis factor alpha, and intercellular adhesion molecule 1 were measured after hypoxia-reoxygenation to assess cell function and injury. Cell morphology, cell viability, and neutrophil adhesion to the CMECs were measured to assess the degree of injury and inflammation. Only cells transfected with Egr-1 antisense oligodeoxyribonucleotide showed a significant reduction in Egr-1 protein expression following hypoxia-reoxygenation. Consistent with the down-regulation of Egr-1 expression, other forms of cell injury were significantly reduced in this group of cells, as evidenced by less alteration in cell morphology, a decrease in expression of tumor necrosis factor alpha and intercellular adhesion molecule 1, improved cell survival, and reduced neutrophil adhesion.


Subject(s)
Coronary Vessels/injuries , Early Growth Response Protein 1/antagonists & inhibitors , Myocardial Reperfusion Injury/prevention & control , Oligodeoxyribonucleotides, Antisense/pharmacology , Animals , Animals, Newborn , Cardiotonic Agents/pharmacology , Cells, Cultured , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cytoprotection/drug effects , Cytoprotection/genetics , Drug Evaluation, Preclinical , Early Growth Response Protein 1/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Microvessels/drug effects , Microvessels/injuries , Microvessels/metabolism , Microvessels/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Rats , Rats, Sprague-Dawley
10.
Oncol Rep ; 24(1): 189-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514461

ABSTRACT

Cell culture, tissue chemistry and flow cytometry were used to determine whether antisense c-Met oligodeoxynucleotides enhanced the sensitivity of human glioma cells to paclitaxel. A combination of paclitaxel with antisense c-Met oligodeoxynucleotides inhibited cell growth, induced apoptosis and induced c-Met protein expression in U251 and SHG44 human glioma cells more significantly than either paclitaxel or the oligodeoxynucleotides on their own (P<0.01). Thus, c-Met antisense oligodeoxynucleotides increase the sensitivity of human glioma cells to paclitaxel. Combined use of the two agents could be a novel and attractive strategy in human glioma treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Glioma/drug therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , Paclitaxel/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Oligodeoxyribonucleotides, Antisense/administration & dosage , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Tumor Cells, Cultured
11.
Pain ; 150(3): 401-413, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573450

ABSTRACT

Anti-nociceptive tolerance to opioids is a well-described phenomenon, which severely limits the clinical efficacy of opioids for the treatment of chronic pain syndromes. The mechanisms that drive anti-nociceptive tolerance, however, are less well understood. We have previously shown that glia have a central role in the development of morphine tolerance and that administration of a glial modulating agent attenuated tolerance formation. Recently, we have demonstrated that morphine enhances microglial Iba1 expression and P2X4 receptor-mediated microglial migration via direct mu opioid receptor signaling in in vitro microglial cultures. We hypothesize that P2X4 receptors drive morphine tolerance and modulate morphine-induced spinal glial reactivity. Additionally, we hypothesize that perivascular microglia play a role in morphine tolerance and that P2X4 receptor expression regulates perivascular microglia ED2 expression. To test these hypotheses, rats were implanted with osmotic minipumps releasing morphine or saline subcutaneously for seven days. Beginning three days prior to morphine treatment, P2X4 receptor antisense oligonucleotide (asODN) was injected intrathecally daily, to selectively inhibit P2X4 receptor expression. P2X4 receptor asODN treatment inhibited morphine-induced P2X4 receptor expression and blocked anti-nociceptive tolerance to systemically administered morphine. P2X4 receptor asODN treatment also attenuated the morphine-dependent increase of spinal ionized calcium binding protein (Iba1), glial fibrillary acidic protein (GFAP) and mu opioid receptor protein expression. Chronic morphine also decreased perivascular microglial ED2 expression, which was reversed by P2X4 receptor asODN. Together, these data suggest that the modulation of P2X4 receptor expression on microglia and perivascular microglia may prove an attractive target for adjuvant therapy to attenuate opioid-induced anti-nociceptive tolerance.


Subject(s)
Calcium-Binding Proteins/metabolism , Drug Tolerance/physiology , Gene Expression Regulation/physiology , Microglia/metabolism , Morphine/administration & dosage , Receptors, Opioid/metabolism , Receptors, Purinergic P2X4/metabolism , Analysis of Variance , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Behavior, Animal/drug effects , CD11b Antigen/metabolism , Disease Models, Animal , Drug Interactions , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Male , Microfilament Proteins , Microglia/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Pain Measurement , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism , Receptors, Purinergic P2X4/genetics , Signal Transduction , Spinal Cord/pathology , Time Factors
12.
J Infect Dis ; 201(12): 1822-30, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20438352

ABSTRACT

BACKGROUND: Members of the Burkholderia cepacia complex (Bcc) cause considerable morbidity and mortality in patients with chronic granulomatous disease and cystic fibrosis. Many Bcc strains are antibiotic resistant, which requires the exploration of novel antimicrobial approaches, including antisense technologies such as phosphorodiamidate morpholino oligomers (PMOs). METHODS: Peptide-conjugated PMOs (PPMOs) were developed to target acpP, which encodes an acyl carrier protein (AcpP) that is thought to be essential for growth. Their antimicrobial activities were tested against different strains of Bcc in vitro and in infection models. RESULTS: PPMOs targeting acpP were bactericidal against clinical isolates of Bcc (>4 log reduction), whereas a PPMO with a scrambled base sequence (scrambled PPMO) had no effect on growth. Human neutrophils were infected with Burkholderia multivorans and treated with AcpP PPMO. AcpP PPMO augmented killing, compared with neutrophils alone and compared with neutrophils alone plus scrambled PPMO. Mice with chronic granulomatous disease that were infected with B. multivorans were treated with AcpP PPMO, scrambled PPMO, or water at 0, 3, and 6 h after infection. Compared with water-treated control mice, the AcpP PPMO-treated mice showed an approximately 80% reduction in the risk of dying by day 30 of the experiment and relatively little pathology. CONCLUSION: AcpP PPMO is active against Bcc infections in vitro and in vivo.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Burkholderia Infections/drug therapy , Burkholderia cepacia complex/drug effects , Morpholines/therapeutic use , Oligodeoxyribonucleotides, Antisense/therapeutic use , Acyl Carrier Protein/antagonists & inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Burkholderia Infections/mortality , Burkholderia Infections/pathology , Cell Survival , Disease Models, Animal , Granulomatous Disease, Chronic/complications , Humans , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Morpholines/pharmacology , Morpholinos , Neutrophils/microbiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Survival Analysis
13.
Synapse ; 64(11): 808-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20340173

ABSTRACT

The 70-kDa mitochondrial heat shock protein, mortalin, is a ubiquitously expressed, multifunctional protein that is capable of binding the neurotransmitter, dopamine, within the brain. Dopamine dysregulation has been implicated in many of the abnormal neurological behaviors. Although studies have indicated that mortalin is differentially regulated in response to dopaminergic modulation, research has yet to elucidate the role of mortalin in the regulation of dopaminergic activity. This study seeks to investigate the role of mortalin in the regulation of dopamine-dependent behavior, specifically as it pertains to schizophrenia (SCZ). Mortalin expression was knocked down through the infusion of antisense oligodeoxynucleotide molecules into the medial prefrontal cortex (mPFC). Rats infused with mortalin antisense oligodeoxynucleotide molecules exhibited significant prepulse inhibition deficits, suggestive of defects in normal sensorimotor gating. Furthermore, mortalin misexpression within the mPFC was coupled to a significant increase in mortalin protein expression within the nucleus accumbens at the molecular level. These findings demonstrate that mortalin plays an essential role in the regulation of dopamine-dependent behavior and plays an even greater role in the pathogenesis of SCZ.


Subject(s)
HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Prefrontal Cortex/metabolism , Reflex, Startle/physiology , Sensory Gating/physiology , Acoustic Stimulation/methods , Animals , Behavior, Animal , Dopamine/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Interpersonal Relations , Male , Oligodeoxyribonucleotides, Antisense/pharmacology , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Sensory Gating/drug effects
14.
Endocrinology ; 149(12): 6416-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18687780

ABSTRACT

Circulating angiotensin II (ANGII) elicits water intake and activates the hypothalamic-pituitary-adrenal (HPA) axis by stimulating angiotensin type 1 receptors (AT1Rs) within circumventricular organs. The subfornical organ (SFO) and the organum vasculosum of the lamina terminalis (OVLT) are circumventricular organs that express AT1Rs that bind blood-borne ANGII and stimulate integrative and effector regions of the brain. The goal of these studies was to determine the contribution of AT1Rs within the SFO and OVLT to the water intake and HPA response to increased circulating ANGII. Antisense oligonucleotides directed against the AT1R [AT1R antisense (AT1R AS)] were administered into the OVLT or SFO. Quantitative receptor autoradiography confirmed that AT1R AS decreased ANGII binding in the SFO and OVLT compared with the scrambled sequence control but did not affect AT1R binding in other nuclei. Subsequently, water intake, ACTH, and corticosterone (CORT) were assessed after administration of isoproterenol, a beta-adrenergic agonist that decreases blood pressure and elevates circulating ANGII. Delivery of AT1R AS into the SFO attenuated water intake, ACTH, and CORT after isoproterenol, whereas similar treatment in the OVLT had no effect. To determine the specificity of this blunted drinking and HPA response, the same parameters were measured after treatment with hypertonic saline, a stimulus that induces drinking independently of ANGII. Delivery of AT1R AS into the SFO or OVLT had no effect on water intake, ACTH, or CORT after hypertonic saline. The results imply that AT1R within the SFO mediate drinking and HPA responses to stimuli that increase circulating ANGII.


Subject(s)
Drinking/drug effects , Hypothalamo-Hypophyseal System/drug effects , Isoproterenol/pharmacology , Pituitary-Adrenal System/drug effects , Receptor, Angiotensin, Type 1/metabolism , Subfornical Organ/drug effects , Adrenergic beta-Agonists/pharmacology , Adrenocorticotropic Hormone/blood , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Oligodeoxyribonucleotides, Antisense/pharmacology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiology , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/physiology , Sodium Chloride/pharmacology , Subfornical Organ/metabolism , Subfornical Organ/physiology
15.
J Neurochem ; 106(5): 2015-29, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18513372

ABSTRACT

Epidermal fatty acid-binding protein (E-FABP), a member of the family of FABPs, exhibits a robust expression in neurons during axonal growth in development and in nerve regeneration following nerve injury. This study examines the impact of E-FABP expression in normal neurite extension in differentiating pheochromocytoma cell (PC12) cultures supplemented with selected long chain free fatty acids (LCFFA). We found that E-FABP binds to a broad range of saturated and unsaturated LCFFAs, including those with potential interest for neuronal differentiation and axonal growth such as C22:6n-3 docosahexaenoic acid (DHA), C20:5n-3 eicosapentaenoic acid (EPA), and C20:4n-6 arachidonic acid (ARA). PC12 cells exposed to nerve growth factor (NGFDPC12) exhibit high E-FABP expression that is blocked by mitogen-activated protein kinase kinase (MEK) inhibitor U0126. Nerve growth factor-differentiated pheochromocytoma cells (NGFDPC12) antisense clones (NGFDPC12-AS) which exhibit low E-FABP expression have fewer/shorter neurites than cells transfected with vector only or NGFDPC12 sense cells (NGFDPC12-S). Replenishing NGFDPC12-AS cells with biotinylated recombinant E-FABP (biotin-E-FABP) protein restores normal neurite outgrowth. Cellular localization of biotin-E-FABP in NGFDPC12 was detected mostly in the cytoplasm and in the nuclear region. Treatment of NGFDPC12 with DHA, EPA, or ARA further enhances neurite length but it does not trigger further induction of TrkA or MEK phosphorylation or E-FABP mRNA observed in differentiating PC12 cells without LCFFA supplementation. Significantly, DHA and EPA neurite stimulating effects are higher in NGFDPC12-S than in NGFDPC12-AS cells. These findings are consistent with the scenario that neurite extension of differentiating PC12 cells, including further stimulation by DHA and EPA, requires sufficient cellular levels of E-FABP.


Subject(s)
Cell Differentiation/physiology , Eye Proteins/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Lipid Metabolism/physiology , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Animals , Cell Compartmentation/physiology , Cell Differentiation/drug effects , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Eye Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Lipid Metabolism/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Neurites/drug effects , Neurites/ultrastructure , Oligodeoxyribonucleotides, Antisense/pharmacology , PC12 Cells , Protein Binding/drug effects , Protein Binding/physiology , Rats , Recombinant Fusion Proteins/pharmacology
16.
Physiol Genomics ; 31(2): 306-14, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17684035

ABSTRACT

Neuropeptide Y (NPY) is an appetite-controlling neuromodulator that contributes to the appetite-suppressing effect of phenylpropanolamine (PPA). Aims of this study were to investigate whether protein kinase A (PKA) signaling is involved in regulating NPY gene expression and PPA-induced anorexia. Rats were given daily with PPA for 5 days. Changes in daily food intake and hypothalamic NPY, PKA, cAMP response element binding protein (CREB), and pro-opiomelanocortin (POMC) gene expression were measured and compared. To further determine if PKA was involved, intracerebroventricular infusions of antisense oligodeoxynucleotide were performed at 60 min before daily PPA treatment in freely moving rats. Results showed that daily PKA, CREB, and POMC expression were increased following PPA treatment, which showed a closely reverse relationship with alterations of decreased feeding behaviors and NPY mRNA levels. Results also showed that PKA knock-down could block PPA-induced anorexia as well as restore NPY mRNA level, indicating the involvement of PKA signaling in the regulation of NPY gene expression. It is suggested that hypothalamic PKA signaling may participate in the central regulation of PPA-mediated appetite suppression via the modulation of hypothalamic NPY gene expression. The present findings reveal that manipulations at the molecular level of PKA or cAMP may allow the development of therapeutic agents to improve the undesirable properties of PPA or other amphetamine-like anorectic drugs.


Subject(s)
Appetite Depressants/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Neuropeptide Y/biosynthesis , Phenylpropanolamine/pharmacology , Animals , Anorexia/chemically induced , Anorexia/genetics , Anorexia/physiopathology , Appetite Depressants/toxicity , Body Weight/drug effects , Cyclic AMP/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Feeding Behavior/physiology , Gene Targeting , Hypothalamus/physiology , Injections, Intraventricular , Male , Neuropeptide Y/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Phenylpropanolamine/toxicity , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , RNA, Messenger/biosynthesis , Rats , Transcription, Genetic
17.
Biochem Biophys Res Commun ; 349(3): 969-75, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16970909

ABSTRACT

TTF-1 is a member of the NKx family of homeodomain genes, and is required for morphogenesis and fetal diencephalon development. Our previous studies have shown that TTF-1 expression is maintained in some regions of the postnatal rat brain and transactivates the gene expression of several neuropeptides. In this study, a potential role for TTF-1 in the regulation of feeding behavior was identified. Immunohistochemical analysis showed that TTF-1 is present in several hypothalamic nuclei of the adult rat brain involved in the control of feeding behavior. Food deprivation for two days markedly increased the hypothalamic levels of TTF-1 mRNA and protein. Intracerebroventricular administration of an antisense TTF-1 oligodeoxynucleotide significantly decreased TTF-1 protein abundance in the hypothalamus. This TTF-1 decrease was followed by a significant decrease in neuropeptide Y mRNA content and an increase in proopiomelanocortin mRNA content, and in turn resulted in a decrease of the animal's food intake and body weight. These results suggest a novel role for TTF-1 in the regulation of feeding behavior in the rat hypothalamus.


Subject(s)
Feeding Behavior , Hypothalamus/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Body Weight/drug effects , Feeding Behavior/drug effects , Hypothalamus/drug effects , Injections, Intravenous , Male , Neuropeptide Y/genetics , Nuclear Proteins/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
18.
Mol Cells ; 22(1): 30-5, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16951547

ABSTRACT

Munc18, a mammalian homolog of C. elegans Unc, is essential for neurotransmitter release. The aim of this study was to identify estrogen-dependent expression of Munc18-1 and its role in the regulation of glutamate release for puberty onset. Hypothalamic munc18-1 mRNA levels were significantly increased by estrogen treatment in ovariectomized, immature female rats. During pubertal development, the munc18-1 mRNA levels dramatically increased between the juvenile period and the anestrous phase of puberty. Intracerebroventricular administration of an antisense oligodeoxynucleotide against munc18-1 mRNA significantly decreased glutamate release and delayed the day of puberty onset. These results suggest that Munc18-1, expressed in an estrogen-dependent manner, plays an important role in the onset of female puberty via the regulation of glutamate release.


Subject(s)
Glutamic Acid/metabolism , Munc18 Proteins/physiology , Animals , Estradiol/pharmacology , Female , Hypothalamus/metabolism , Munc18 Proteins/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Ovariectomy , Ovary/physiology , Rats
19.
Pain ; 123(1-2): 75-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16545521

ABSTRACT

Antisense (AS) oligodeoxynucleotides (ODNs) targeting the Nav 1.8 sodium channel have been reported to decrease inflammatory hyperalgesia and L5/L6 spinal nerve ligation-induced mechanical allodynia in rats. The present studies were conducted to further characterize Nav 1.8 AS antinociceptive profile in rats to better understand the role of Nav 1.8 in different pain states. Consistent with earlier reports, chronic intrathecal Nav 1.8 AS, but not mismatch (MM), ODN decreased TTX-resistant sodium current density (by 60.5+/-10.2% relative to MM; p<0.05) in neurons from L4 to L5 dorsal root ganglia and significantly attenuated mechanical allodynia following intraplantar complete Freund's adjuvant. In addition, 10 days following chronic constriction injury of the sciatic nerve, Nav 1.8 AS, but not MM, ODN also attenuated mechanical allodynia (54.3+/-8.2% effect, p<0.05 vs. MM) 2 days after initiation of ODN treatment. The anti-allodynic effects remained for the duration of the AS treatment, and CCI rats returned to an allodynic state 4 days after discontinuing AS. In contrast, Nav 1.8 AS ODN failed to reduce mechanical allodynia in the vincristine chemotherapy-induced neuropathic pain model or a skin-incision model of post-operative pain. Finally, Nav 1.8 AS, but not MM, ODN treatment produced a small but significant attenuation of acute noxious mechanical sensitivity in naïve animals (17.6+/-6.2% effect, p<0.05 vs. MM). These data demonstrate a greater involvement of Nav 1.8 in frank nerve injury and inflammatory pain as compared to acute, post-operative or chemotherapy-induced neuropathic pain states.


Subject(s)
Hyperalgesia/physiopathology , Inflammation/physiopathology , Nerve Tissue Proteins/physiology , Neuralgia/physiopathology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Pain, Postoperative/physiopathology , Sodium Channels/physiology , Vincristine/toxicity , Animals , Behavior, Animal , Drug Evaluation, Preclinical , Freund's Adjuvant/toxicity , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Injections, Spinal , Ion Transport , Ligation , Male , NAV1.8 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Neuralgia/chemically induced , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Pressure/adverse effects , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Sodium/physiology , Sodium Channels/drug effects , Sodium Channels/genetics , Spinal Nerves/injuries , Stress, Mechanical , Tetrodotoxin/pharmacology
20.
Toxicol Appl Pharmacol ; 212(2): 99-109, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16084549

ABSTRACT

Amphetamine (AMPH) is known as an anorectic agent. The mechanism underlying the anorectic action of AMPH has been attributed to its inhibitory action on hypothalamic neuropeptide Y (NPY), an appetite stimulant in the brain. This study was aimed to examine the molecular mechanisms behind the anorectic effect of AMPH. Results showed that AMPH treatment decreased food intake, which was correlated with changes of NPY mRNA level, but increased c-fos, c-jun and superoxide dismutase (SOD) mRNA levels in hypothalamus. To determine if c-fos or c-jun was involved in the anorectic response of AMPH, infusions of antisense oligonucleotide into the brain were performed at 1 h before daily AMPH treatment in freely moving rats, and the results showed that c-fos or c-jun knockdown could block this anorectic response and restore NPY mRNA level. Moreover, c-fos or c-jun knockdown could partially block SOD mRNA level that might involve in the modulation of NPY gene expression. It was suggested that c-fos/c-jun signaling might involve in the central regulation of AMPH-mediated feeding suppression via the modulation of NPY gene expression.


Subject(s)
Amphetamine/pharmacology , Appetite Depressants/pharmacology , Genes, fos/physiology , Genes, jun/physiology , Hypothalamus/metabolism , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Animals , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation/drug effects , Hypothalamus/enzymology , Injections, Intraventricular , Male , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL