Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Inflammopharmacology ; 32(2): 1421-1437, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37541971

ABSTRACT

Parkinson's disease (PD) is the second most common progressive age-related neurodegenerative disorder. Paramount evidence shed light on the role of PI3K/AKT signaling activation in the treatment of neurodegenerative disorders. PI3K/AKT signaling can be activated via cAMP-dependent pathways achieved by phosphodiesterase 4 (PDE4) inhibition. Roflumilast is a well-known PDE4 inhibitor that is currently used in the treatment of chronic obstructive pulmonary disease. Furthermore, roflumilast has been proposed as a favorable candidate for the treatment of neurological disorders. The current study aimed to unravel the neuroprotective role of roflumilast in the rotenone model of PD in rats. Ninety male rats were allocated into six groups as follows: control, rotenone (1.5 mg/kg/48 h, s.c.), L-dopa (22.5 mg/kg, p.o), and roflumilast (0.2, 0.4 or 0.8 mg/kg, p.o). All treatments were administrated for 21 days 1 h after rotenone injection. Rats treated with roflumilast showed an improvement in motor activity and coordination as well as preservation of dopaminergic neurons in the striatum. Moreover, roflumilast increased cAMP level and activated the PI3K/AKT axis via stimulation of CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling cascades. Roflumilast also caused an upsurge in mTOR and Nrf2, halted GSK-3ß and NF-ĸB, and suppressed FoxO1 and caspase-3. Our study revealed that roflumilast exerted neuroprotective effects in rotenone-induced neurotoxicity in rats. These neuroprotective effects were mediated via the crosstalk between CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling pathways which activates PI3K/AKT trajectory. Therefore, PDE4 inhibition is likely to offer a reliable persuasive avenue in curing PD via PI3K/AKT signaling activation.


Subject(s)
Aminopyridines , Benzamides , Neuroprotective Agents , Parkinson Disease , Animals , Male , Rats , Brain-Derived Neurotrophic Factor/metabolism , Cyclopropanes , Glycogen Synthase Kinase 3 beta , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rotenone , Sirtuin 1
2.
Neurochem Res ; 48(11): 3296-3315, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37493882

ABSTRACT

Hot peppers, also called chilli, chilli pepper, or paprika of the plant genus Capsicum (family Solanaceae), are one of the most used vegetables and spices worldwide. Capsaicin (8-methyl N-vanillyl-6-noneamide) is the main pungent principle of hot green and red peppers. By acting on the capsaicin receptor or transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1), capsaicin selectively stimulates and in high doses defunctionalizes capsaicin-sensitive chemonociceptors with C and Aδ afferent fibers. This channel, which is involved in a wide range of neuronal processes, is expressed in peripheral and central branches of capsaicin-sensitive nociceptive neurons, sensory ganglia, the spinal cord, and different brain regions in neuronal cell bodies, dendrites, astrocytes, and pericytes. Several experimental and clinical studies provided evidence that capsaicin protected against ischaemic or excitotoxic cerebral neuronal injury and may lower the risk of cerebral stroke. By preventing neuronal death, memory impairment and inhibiting the amyloidogenic process, capsaicin may also be beneficial in neurodegenerative disorders such as Parkinson's or Alzheimer's diseases. Capsaicin given in systemic inflammation/sepsis exerted beneficial antioxidant and anti-inflammatory effects while defunctionalization of capsaicin-sensitive vagal afferents has been demonstrated to increase brain oxidative stress. Capsaicin may act in the periphery via the vagal sensory fibers expressing TRPV1 receptors to reduce immune oxidative and inflammatory signalling to the brain. Capsaicin given in small doses has also been reported to inhibit the experimentally-induced epileptic seizures. The aim of this review is to provide a concise account on the most recent findings related to this topic. We attempted to delineate such mechanisms by which capsaicin exerts its neuronal protective effects. We also aimed to provide the reader with the current knowledge on the mechanism of action of capsaicin on sensory receptors.


Subject(s)
Capsaicin , TRPV Cation Channels , Capsaicin/pharmacology , Capsaicin/therapeutic use , TRPV Cation Channels/metabolism , Neuroprotection , Nociceptors/metabolism , Spinal Cord/metabolism , Gonadal Steroid Hormones
3.
Neurochem Res ; 45(5): 1045-1061, 2020 May.
Article in English | MEDLINE | ID: mdl-32036609

ABSTRACT

The transient receptor potential vanilloid-1 (TRPV1) receptor has been implicated in the development of epileptic seizures. We examined the effect of the TRPV1 agonist capsaicin on epileptic seizures, neuronal injury and oxidative stress in a model of status epilepticus induced in the rat by intraperitoneal (i.p.) injections of pentylenetetrazole (PTZ). Capsaicin was i.p. given at 1 or 2 mg/kg, 30 min before the first PTZ injection. Other groups were i.p. treated with the vehicle or the anti-epileptic drug phenytoin (30 mg/kg) alone or co-administered with capsaicin at 2 mg/kg. Brain levels of malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide, and paraoxonase-1 (PON-1) activity, seizure scores, latency time and PTZ dose required to reach status epilepticus were determined. Histopathological assessment of neuronal damage was done. Results showed that brain MDA decreased by treatment with capsaicin, phenytoin or capsaicin/phenytoin. Nitric oxide decreased by capsaicin or capsaicin/phenytoin. GSH and PON-1 activity increased after capsaicin, phenytoin or capsaicin/phenytoin. Mean total seizure score decreased by 48.8% and 66.3% by capsaicin compared with 78.7% for phenytoin and 69.8% for capsaicin/phenytoin treatment. Only phenytoin increased the latency (115.7%) and threshold dose of PTZ (78.3%). Capsaicin did not decrease the anti-convulsive effect of phenytoin but prevented the phenytoin-induced increase in latency time and threshold dose. Neuronal damage decreased by phenytoin or capsaicin at 2 mg/kg but almost completely prevented by capsaicin/phenytoin. Thus in this model of status epilepticus, capsaicin decreased brain oxidative stress, the severity of seizures and neuronal injury and its co-administration with phenytoin afforded neuronal protection.


Subject(s)
Anticonvulsants/therapeutic use , Capsaicin/therapeutic use , Neuroprotective Agents/therapeutic use , Pentylenetetrazole/toxicity , Seizures/chemically induced , Seizures/prevention & control , Animals , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Seizures/metabolism
4.
Neurochem Res ; 45(5): 1062-1063, 2020 May.
Article in English | MEDLINE | ID: mdl-32100166

ABSTRACT

The original version of this article unfortunately contains an error in the Y axis units in Fig. 1b, c (the symbol µ is not clear: µmol/g.tissue). This has been corrected by publishing this erratum.

5.
J Nanosci Nanotechnol ; 18(1): 564-575, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29768882

ABSTRACT

The aim of this study was to investigate the effect of treatment with grape seed extract (GSE) on the neurotoxic and genotoxic effects of acute malathion exposure. Rats received malathion (150 mg/kg by i.p. injection) for two successive days alone or combined with GSE at doses of 150 or 300 mg/kg, orally or with GSE at 300 mg/kg and atropine at a dose of 2 mg/kg, i.p. Malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide, paraoxonase (PON1) were determined in cortex, striatum, and rest of brain tissue (subcortex). Interleukin-1ß (IL-1ß), and butyrylcholinesterase (BChE) activities were determined in brain regions. Cytogenetic analyses for chromosomal aberrations in somatic and germ cells, micronucleus test, Comet assay, DNA fragmentation of liver cells and histopathological examination of brain and liver sections were also performed. Malathion resulted in an increase in MDA, nitric oxide; a decrease in GSH and PON1 activity in different brain regions. IL-1ß increased, while BChE activity decreased in brain after the administration of malathion. The insecticide also caused marked structural and numerical chromosomal aberrations and increased liver DNA fragmentation. The Comet assay showed a significant increase in DNA damage of peripheral blood lymphocytes. These effects of malathion were alleviated with the administration of GSE alone or combined with atropine. Addition of atropine to treatment with GSE was associated with significant decrease in MDA, BChE and chromosomal aberrations compared with GSE only treatment. Our data indicate that GSE protects against malathion neurotoxic and genotoxic effects, most likely through reducing brain oxidative stress and inflammatory response.


Subject(s)
Grape Seed Extract , Malathion , Mutagens , Animals , Antioxidants , Atropine , DNA Damage , Grape Seed Extract/pharmacology , Malathion/toxicity , Malondialdehyde , Mutagens/toxicity , Neurotoxins/toxicity , Oxidative Stress , Rats
6.
J Nanosci Nanotechnol ; 18(1): 510-521, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29768876

ABSTRACT

We examined the effect of treatment with neostigmine alone or with atropine on brain oxidative stress and on brain and liver tissue damage following acute malathion toxicity. Rats were intraperitoneally treated with malathion 150 mg/kg along with neostigmine (200 or 400 µg/kg) or neostigmine (200 µg/kg) + atropine (1 mg/kg) and euthanized 4 h later. Results indicated that compared with the saline group, malathion resulted in (i) higher brain malondialdehyde (MDA) and nitric oxide (46.4% and 86.2%); (ii) decreased brain reduced glutathione (GSH) (67.6%); (iii) decreased brain paraoxonase-1 (PON1), acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activities (31.2%, 21.6% and 60%); (iv) decreased brain glucose (-38.1%); (v) neuronal degeneration in cortex and hippocampus and markedly increased glial fibrillary acidic protein (GFAP) immunostaining in the hippocampus; (v) hydropic and fatty degeneration in liver. Rats treated with malathion along with neostigmine or neostigmine + atropine showed no change in brain MDA but decreased nitric oxide (-34.2%-48%). GSH increased after neostigmine 200 µg/kg or neostigmine + atropine (35.8% and 41%). PON1 activity increased (42%-35.2%) and glucose concentrations increased (91.5%-81.5%) by 400 µg/kg neostigmine or neostigmine + atropine. Brain AChE activity remained unchanged but BChE activity showed 18.3% increment after 400 µg/kg neostigmine. Rats treated with 400 µg/kg neostigmine or neostigmine + atropine had normal neuronal appearance in cortex and hippocampus and weak GFAP expression in hippocampus. Liver damage was prevented by neostigmine + atropine. These results suggest that treatment with neostigmine + atropine afforded protection against the deleterious effects of acute malathion on the brain and liver.


Subject(s)
Malathion , Neostigmine , Animals , Atropine/pharmacology , Brain/drug effects , Liver/drug effects , Malathion/toxicity , Neostigmine/pharmacology , Oxidative Stress , Rats
7.
J Neural Transm (Vienna) ; 120(12): 1673-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23794033

ABSTRACT

This study investigated the role of vagal innervation in oxidative stress after systemic administration of lipopolysaccharide (LPS) endotoxin. Control rats and rats subjected to bilateral subdiaphragmatic vagotomy, perivagal capsaicin application (5 mg/ml) or cholinergic receptor blockade with subcutaneous atropine (1 mg/kg), were intraperitoneally injected with 300 µg/kg of LPS and euthanized 4 h later. Results indicated that; (1) surgical vagotomy and sensory denervation by perivagal capsaicin increased brain oxidative stress and decreased reduced glutathione in basal condition (saline-treated rats) and following endotoxin challenge; (2) oxidative stress decreased after cholinergic blockade with atropine in endotoxemic rats; (3) nitric oxide decreased by abdominal vagotomy, sensory deafferentation and cholinergic blockade after endotoxin injection; (4) liver lipid peroxidation decreased after surgical vagotomy and cholinergic blockade but increased after sensory deafferentation; (5) liver reduced glutathione decreased following vagotomy and sensory denervation in basal state and by cholinergic blockade in basal state and during endotoxemia; (6) nitric oxide increased by vagotomy in basal state and by sensory denervation and cholinergic blockade in basal state and during endotoxemia; (7) liver histological damage increased by subdiaphragmatic vagotomy, sensory denervation or cholinergic blockade. These findings suggest that: (1) sensory fibers (signals from the periphery) running in the vagus nerves are important in maintaining the redox status of the brain; (2) capsaicin vagal sensory nerves are likely to maintain nitric oxide tone in basal conditions; (3) the vagus nerve modulates liver redox status and nitric oxide release, (4) the vagus nerve mediates protective role in the liver with both cholinergic and capsaicin-sensitive mechanisms being involved.


Subject(s)
Endotoxins/pharmacology , Lipopolysaccharides/pharmacology , Oxidative Stress/drug effects , Vagotomy , Vagus Nerve/physiology , Afferent Pathways/drug effects , Afferent Pathways/physiology , Animals , Atropine/pharmacology , Brain/drug effects , Brain/metabolism , Capsaicin/pharmacology , Efferent Pathways/drug effects , Efferent Pathways/physiology , Female , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Malondialdehyde/metabolism , Muscarinic Antagonists/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley
8.
Iran J Basic Med Sci ; 26(3): 335-342, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36865040

ABSTRACT

Objectives: This study investigated the therapeutic effect of red hot pepper (Capsicum annuum) methanolic extract in induced Alzheimer's disease using AlCl3 in male rats. Materials and Methods: Rats were injected with AlCl3 intraperitoneally (IP) daily for two months. Starting from the 2nd month of AlCl3, rats received, in addition, IP treatments with Capsicum extract (25 and 50 mg/kg) or saline. Other groups received only saline or Capsicum extract at 50 mg/kg for two months. Brain levels of reduced glutathione (GSH), nitric oxide (NO), and malondialdehyde (MDA) were determined. Additionally, paraoxonase-1 (PON-1) activity, interleukin-6 (IL-6), Aß-peptide, and acetylcholinesterase (AChE) concentrations in the brain were measured. Behavioral testing included wire-hanging tests for neuromuscular strength and memory tests such as Y-maze and Morris water maze. Histopathology of the brain was also done. Results: Compared with saline-treated rats, AlCl3 caused significant elevation of brain oxidative stress as GSH level and PON-1 activity were depleted along with MDA and NO level elevation in the brain. There were also significant increases in brain Aß-peptide, IL-6, and AChE levels. Behavioral testing indicated that AlCl3 decreased neuromuscular strength and impaired memory performance. Capsicum extract given to AlCl3-treated rats significantly alleviated oxidative stress and decreased Aß-peptide and IL-6 in the brain. It also improved grip strength and memory functioning and prevented neuronal degeneration in the cerebral cortex, hippocampus, and substantia nigra of AlCl3-treated rats. Conclusion: Short-term administration of ASA (50 mg/kg) has adverse effects on male reproductive function in mice. Co-administration of melatonin protects against ASA-induced impairment of male reproductive function by preventing the reduction in serum TAC and testosterone levels seen with ASA treatment alone.

9.
Inflammopharmacology ; 20(4): 207-17, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22127606

ABSTRACT

This study investigated the effect of capsaicin (the active principle of hot red pepper and a sensory excitotoxin) on oxidative stress after systemic administration of the endotoxin lipopolysaccharide (100 µg/kg, i.p.) in rats. Capsaicin (15, 150 or 1,500 µg/kg; 10, 100 or 400 µg/mL) was given via intragastric (i.g.) or intraperitoneal (i.p.) routes at time of endotoxin administration. Rats were killed 4 h later. Malondialdehyde (MDA) and reduced glutathione (GSH) were measured in brain, liver, and lungs. Alanine aminotransferase (ALT), aspartate aminotransferase, alkaline phosphatase (ALP), nitric oxide, and glucose were measured in serum. In addition, histopathological examination of liver tissue was performed. In LPS-treated rats, hepatic GSH increased significantly by 40.8% after i.p. capsaicin at 1,500 µg/kg. Liver MDA increased significantly by 32.9% after the administration of i.g. capsaicin at 1,500 µg/kg and by 27.8 and 37.6% after the administration of i.p. capsaicin at 150 and 1,500 µg/kg, respectively. In lung tissue, both MDA and GSH were decreased by capsaicin administration. MDA decreased by 19-20.8% after i.g. capsaicin and by 17.5-23.2% after i.p. capsaicin (150-1,500 µg/kg), respectively. GSH decreased by 39.3-64.3% and by 35.7-41.1% after i.g. or i.p. capsaicin (150-1,500 µg/kg), respectively. Brain GSH increased significantly after the highest dose of i.g. or i.p. capsaicin (by 20.6 and 15.9%, respectively). The increase in serum ALT and ALP after endotoxin administration was decreased by oral or i.p. capsaicin. Serum nitric oxide showed marked increase after LPS injection, but was markedly decreased after capsaicin (1,500 µg/kg, i.p.). Serum glucose increased markedly after the administration of LPS, and was normalized by capsaicin treatment. It is suggested that in the presence of mild systemic inflammation, acute capsaicin administration might alter oxidative status in some tissues and exert an anti-inflammatory effect. Capsaicin exerted protective effects in the liver and lung against the LPS-induced tissue damage.


Subject(s)
Capsaicin/pharmacology , Lipopolysaccharides/toxicity , Oxidative Stress/drug effects , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Body Temperature/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Lung/metabolism , Nitric Oxide/blood , Rats , Rats, Sprague-Dawley
10.
Article in English | MEDLINE | ID: mdl-23089532

ABSTRACT

BACKGROUND: Gabapentin, a structural analog of γ-aminobutyric acid (GABA), is used in the treatment of neuropathic pain in multiple sclerosis. METHODS: This study investigated the effect of gabapentin on oxidative stress in a model of brain demyelination evoked by intracerebral injection (i.c.i) of ethidium bromide (10 µL of 0.1%). Rats received saline (control) or gabapentin at 100 or 300 mg/kg orally daily for 10 days prior to injection of ethidium bromide. Rats were euthanized 1 day later, and then the levels of reduced glutathione (GSH), glutathione peroxidase (GPx) activity, lipid peroxidation (malondialdehyde; MDA), nitrite, acetyl cholinesterase (AChE) and paraoxonase activities were assessed in the brain cortex in different treatment groups. RESULTS: Ethidium bromide resulted in increased oxidative stress in the cortex 1 day after its injection. Malondialdehyde increased by 30.2%, whereas GSH decreased by 17.6%. GPx activity was inhibited by 78.6%. Brain nitrite increased by 55.4%, AChE activity decreased by 33.4% and paraoxonase activity decreased by 27.5%. In ethidium bromide treated rats, gabapentin administered at 300 mg/kg increased cortical MDA by 66%. GSH was unaltered by gabapentin, but GPx activity decreased by 54.3% by the higher dose of gabapentin. Nitrite decreased by 21.4% and 29.2% after 100 and 300 mg/kg of gabapentin, respectively. AChE activity increased by 28.6% and 69.3% by 100 and 300 mg/kg of gabapentin, respectively. Paraoxonase activity showed 83.3% and 73% decreases by 100 and 300 mg/kg of gabapentin, respectively. CONCLUSIONS: These results suggest that gabapentin increases brain lipid peroxidation and decreases brain antioxidant enzymes in this model of chemical demyelination.


Subject(s)
Amines/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Demyelinating Diseases/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Oxidative Stress/drug effects , gamma-Aminobutyric Acid/pharmacology , Amines/administration & dosage , Animals , Antioxidants/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclohexanecarboxylic Acids/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Ethidium/toxicity , Excitatory Amino Acid Antagonists/administration & dosage , Gabapentin , Lipid Peroxidation/drug effects , Male , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/administration & dosage
11.
Neurochem Res ; 36(6): 1062-72, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21448596

ABSTRACT

UNLABELLED: We studied the role of oxidative stress and the effect of vinpocetine (1.5, 3 or 6 mg/kg) and piracetam (150 or 300 mg/kg) in acute demyelination of the rat brain following intracerebral injection of ethidium bromide (10 µl of 0.1%). RESULTS: ethidium bromide caused (1) increased malondialdehyde (MDA) in cortex, hippocampus and striatum; (2) decreased total antioxidant capacity (TAC) in cortex, hippocampus and striatum; (3) decreased reduced glutathione (GSH) in cortex and hippocampus (4); increased serum nitric oxide and (5) increased striatal (but not cortical or hippocampal) acetylcholinesterase (AChE) activity. MDA decreased in striatum and cortex by the lower doses of vinpocetine or piracetam but increased in cortex and hippocampus and in cortex, hypothalamus and striatum by the higher dose of vinpocetine or piracetam, respectively along with decreased TAC. GSH increased by the higher dose of piracetam and by vinpocetine which also decreased serum nitric oxide. Vinpocetine and piracetam displayed variable effects on regional AChE activity.


Subject(s)
Brain/drug effects , Models, Biological , Oxidative Stress , Piracetam/pharmacology , Vinca Alkaloids/pharmacology , Animals , Brain/physiopathology , Male , Rats , Rats, Sprague-Dawley
12.
Iran J Basic Med Sci ; 23(9): 1207-1217, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32963743

ABSTRACT

OBJECTIVES: The effects of low dose amphetamine on oxidative stress and rotenone-induced neurotoxicity and liver injury were examined in vivo in a mice model of Parkinson's disease. MATERIALS AND METHODS: Male mice were treated with rotenone (1.5 mg/kg, every other day for two weeks, subcutaneously). Mice received either the vehicle or amphetamine intraperitoneally at doses of 0.5, 1.0, or 2.0 mg/kg. Oxidative stress was assessed by measurement of the lipid peroxidation product malondialdehyde (MDA), nitric oxide (NO), total anti-oxidant capacity (TAC), and paraoxonase-1 (PON-1) activity in the brain and liver. In addition, brain concentrations of nuclear factor kappa B (NF-κB) and tyrosine hydroxylase were determined and histopathology and Bax/Bcl-2 immunohistochemistry were performed. RESULTS: The levels of lipid peroxidation and NO were increased and TAC and PON-1 were decreased significantly compared with vehicle-injected control mice. There were also significantly increased NF-κB and decreased tyrosine hydroxylase in the brain following rotenone administration. These changes were significantly attenuated by amphetamine. Rotenone caused neurodegenerative changes in the substantia nigra, cerebral cortex, and hippocampus. The liver showed degenerative changes in hepatocytes and infiltration of Kupffer cells. Bax/Bcl2 ratio was significantly increased in brain and liver tissues. Amphetamine prevented these histopathological changes and the increase in apoptosis evoked by rotenone. CONCLUSION: These results suggest that low dose amphetamine exerts anti-oxidant and anti-apoptotic effects, protects against rotenone-induced neurodegeneration, and could prevent neuronal cell degeneration in Parkinson's disease.

13.
Life Sci ; 257: 118070, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32668327

ABSTRACT

AIMS: Several studies suggested that ATP-sensitive potassium channels (KATP) are potential therapeutic targets for protection against various neurodegenerative disorders, yet, there is an ongoing controversy regarding their role in Parkinson's disease (PD). Thus, the aim of the current study is to investigate the protective effect of KATP blockade and activation in the mice rotenone model of PD. MAIN METHODS: PD has been induced by 9 subcutaneous injections of rotenone (1.5 mg/kg; 3 times/week) in adult male Swiss albino mice. For 3 consecutive weeks, parkinsonian mice were either untreated or treated with L-dopa (25 mg/kg), the KATP channel blocker glibenclamide (3 mg/kg) or the KATP channel opener nicorandil (6 mg/kg). KEY FINDINGS: Glibenclamide significantly improved motor performance in the wire hanging and stair tests and halted the decline in striatal dopamine content as well as dopaminergic neurons' density. In addition, it reduced the rotenone-induced apoptosis as portrayed in the immunohistopathological examination via increasing Bcl-2 and decreasing caspases-3, -8, -9 contents. Furthermore, through its anti-inflammatory potential, glibenclamide reduced tumor necrosis factor-alpha level. On the other hand, nicorandil failed to mitigate the rotenone-induced neurodegenerative consequences. SIGNIFICANCE: KATP channel blockade by glibenclamide has neuroprotective effect against rotenone-induced neurotoxicity, that was mediated by its anti-inflammatory effect along with hindering apoptosis through extrinsic and intrinsic pathways.


Subject(s)
Glyburide/pharmacology , KATP Channels/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Potassium Channel Blockers/pharmacology , Animals , Apoptosis/drug effects , Dopamine/metabolism , Dopaminergic Neurons/metabolism , KATP Channels/metabolism , Levodopa/pharmacology , Male , Mice , Nicorandil/pharmacology , Parkinsonian Disorders/physiopathology , Rotenone/toxicity
14.
Prog Drug Res ; 68: v-vi, 2014.
Article in English | MEDLINE | ID: mdl-24941662
15.
Inflammopharmacology ; 17(3): 113-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19557311

ABSTRACT

BACKGROUND: The plant origin capsaicinoids (capsaicin, dihydrocapsaicin, norcapsaicin, dihydrocapsaicin, homocapsaicin, homodihydrocapsaicin) are well known and used as nutritional additive agents in the every day nutritional practice from the last 9,500 years; however, we had have a very little scientifically based knowledge on their chemistry, physiology and pharmacology in animal observations, and in humans up to the mid-twentieth century. Our knowledge about their chemistry, physiology, pharmacology entered to be scientifically based evidence from the year 1980, dominantly in animal observations. The human observations with capsaicin (capsaicinoids), in terms of good clinical practice, have been started only in the last 10-year period (from 1997) in randomized, prospective, multiclinical studies. The name of "capsaicin" used only in the physiological and pharmacological research both in animal experiments and in human observation. The "capsaicin" (as a "chemically" used natural compound) modifies the so-called capsaicin-sensitive afferent nerves depending on their applied doses. AIMS: The specific action of capsaicin (capsaicinoids) on sensory afferent nerves modifying gastrointestinal (GI) function (under very specific conditions) offers a possibility for the production of an orally applicable drug or for other drug combinations, which can be used in the human medical therapy. The production of new drug is based on the critical interdisciplinary review of the results obtained with capsaicinoids. MATERIALS AND METHODS: This paper gives an interdisciplinary and critical overview on the chemical, physiological, pharmacological and toxicological actions of the natural origin capsaicinoids (from the point of drug production) under conditions of acute, subacute and chronic administration in animal experiments and human observations, toxicology, pharmacokinetics). This interdisciplinary review covers the following main chapters: (1) physiological and pharmacological research tool by capsaicin in the animals and human beings, (2) capsaicin research in animals (including the acute, subacute toxicology and chronic toxicology metabolism, genotoxicology), (3) capsaicin observation with capsaicin in human beings. CONCLUSION: (1) The capsaicin used in the physiological and pharmacological observations (in animals and human beings) chemically represents different chemical compounds, which can be obtained from the plants (paprika, chilli, etc.), (2) capsaicinoids are able to modify the capsaicin-sensitive afferent nerves, which have principle roles in the defence of different organs (including the gastrointestinal tract [against the different chemicals, heat, strech, chemical millieu-induced damage], (3) the application of capsaicin (capsaicinoids) can be repeated for the beneficial effects on the gastrointestinal tract as those in animal experiments. After this interdisciplinary and critical review, this paper demonstrates the well-planned research pathways of the discoveries of capsaicinoids from plant chemistry, via physiology, pharmacology and toxicology in animal experiments and human observations.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Capsaicin , Gastrointestinal Diseases/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Capsaicin/therapeutic use , Capsaicin/toxicity , Drug Evaluation, Preclinical , Drug Interactions , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/prevention & control , Humans , Models, Biological , Molecular Structure , Sensory Receptor Cells/drug effects , TRPV Cation Channels/physiology
16.
Neurotoxicology ; 67: 245-258, 2018 07.
Article in English | MEDLINE | ID: mdl-29906478

ABSTRACT

Nicotine and tramadol concomitant drug dependence pose increasing social, economic as well as public threats. Accordingly, the present study investigated neurochemical, neurobehavioral and neuropathological changes in the brain subsequent to the interaction of nicotine and tramadol. To this end, tramadol (20 mg/kg, i.p) and nicotine (0.25 mg/kg, i.p) were administrated to male albino mice once daily for 30 days. Consequent to microglial activation, nicotine exacerbated oxidative/nitrosative stress induced by tramadol as manifest by the step-up in thiobarbituric acid reactive substances and nitric oxide subsequent to the enhanced levels of neuronal and inducible nitric oxide synthases; paralleled by decreased non-protein sulfhydryls. Increased oxidative stress by tramadol and/or nicotine sequentially augmented nuclear factor kappa B and the proinflammatory cytokine tumor necrosis factor α with the induction of apoptosis evident by the increased caspase-3 immunoreactivity. However, paradoxical to the boosted inflammation and apoptosis, heightened DA levels in the cortex parallel along with increased tyrosine hydroxylase in midbrain were apparent. Concomitant administration of tramadol and nicotine impaired spatial navigation in the Morris Water Maze test coupled with enhanced levels of acetyl- and butyryl cholinestrases. However, tramadol in association with nicotine improved social interaction while decreasing anxiety and aggression linked to chronic administration of nicotine, effects manifested by increased levels of serotonin and GABA. These results provide evidence that co-administration of tramadol and nicotine may enhance reward and dependence while reducing anxiety and aggression linked to nicotine administration. However, such combination exacerbated neurotoxic effects and elicited negative effects regarding learning and memory.


Subject(s)
Analgesics, Opioid/administration & dosage , Brain/metabolism , Inflammation Mediators/metabolism , Nicotine/administration & dosage , Opioid-Related Disorders/metabolism , Tramadol/administration & dosage , Analgesics, Opioid/toxicity , Animals , Brain/drug effects , Drug Therapy, Combination , Inflammation Mediators/antagonists & inhibitors , Interpersonal Relations , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Mice , Motor Activity/drug effects , Motor Activity/physiology , Nicotinic Agonists/administration & dosage , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/psychology , Tramadol/toxicity
17.
Eur J Med Chem ; 42(10): 1285-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17408809

ABSTRACT

This study aimed at evaluation of the reactivity of melatonin (1) towards various chemical reagents to produce new melatonin analogues containing heterocyclic moieties which would provide basic pharmacological activities. The pyrrolo[1,2-a]indole derivatives 3, 5, 12, 14 and pyrido[1,2-a]indole derivatives 9a, b were synthesized via straightforward and efficient methods and their structures were established based on the analytical and spectral data. Also, this work was extended to study the potential role of the novel synthesized melatonin analogues 3, 5, 9a and 12 as anti-inflammatory and anti-nociceptive agents in comparison with melatonin. After s.c. administration all compounds induced significant anti-inflammatory activity, inhibiting the paw oedema response compared with the control group at all time points in the test. Compound 5 has the strongest anti-inflammatory activity which exceeds that of the parent reference, melatonin, followed by compounds 9a and 12, at the first 2h of administration. Effect of melatonin analogues on thermal pain, acetic acid-induced writhing and gastric lesions caused by indomethacin was also investigated. Compounds 5 and 12 were more potent as anti-nociceptive drugs; they are more potent in this respect than melatonin.


Subject(s)
Analgesics, Non-Narcotic/chemical synthesis , Analgesics, Non-Narcotic/therapeutic use , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/therapeutic use , Melatonin/analogs & derivatives , Melatonin/therapeutic use , Analgesics, Non-Narcotic/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Edema/drug therapy , Edema/pathology , Extremities , Melatonin/chemical synthesis , Melatonin/chemistry , Mice , Molecular Structure , Rats
18.
ScientificWorldJournal ; 7: 1242-55, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17721639

ABSTRACT

The effect of biphenyldimethyldicarboxylate (DDB), a synthetic compound, in use for the treatment of chronic hepatitis was studied on hepatic injury caused in rats by administration of carbon tetrachloride (CCl4). Starting at time of administration of the first dose of CCl4, rats received DDB at four dose levels (3, 15, 75 or 375 mg/kg), silymarin (22 mg/kg), a combination of DDB (75 mg/kg) and silymarin (22 mg/kg) or saline (control) once orally daily for 30 days. The administration of DDB in CCl4-treated rats at 75 or 375 mg/kg resulted in 61.2-76.2% decrease in alanine aminotransferase (ALT) and 46.9-60.8% decrease in aspartate aminotransferase (AST), respectively compared with the CCl4 control group. Silymarin treatment resulted in 34.6 and 30% decrease in ALT and AST, while DDB (75 mg/kg) combined with silymarin (22 mg/kg) resulted in 58.2 and 31% decrease in ALT and AST, respectively. Serum creatinine increased by 50% by DDB at 375 mg/kg. After treatment with DDB at 75 or 375 mg/kg or DDB combined with silymarin, the development of liver necrosis and fibrosis caused by CCl4 was markedly reduced, while after DDB combined with silymarin no DNA aneuploid cells could be observed. The decrease in glycogen and protein contents in hepatocytes caused by CCl4 was markedly prevented by co-treatment with DDB at 75 or 375 mg/kg or DDB combined with silymarin. It is concluded that in the model of hepatic injury caused by chronic administration of CCl4 in rats, the synthetic compound DDB, limits hepatocellular injury and exerts antifibrotic effect. Better improvement in protein, DNA, mucopolysaccharide content was seen after both DDB and silymarin compared to DDB alone. It is suggested, therefore, that DDB alone or in combination with silymarin might prove of benefit in the therapy of chronic liver disease. Monitoring of kidney functions in patients taking DDB is warranted.


Subject(s)
Carbon Tetrachloride , Dioxoles/administration & dosage , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/metabolism , Silymarin/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antioxidants/administration & dosage , Dose-Response Relationship, Drug , Drug Therapy, Combination , Liver Cirrhosis, Experimental/chemically induced , Rats , Rats, Sprague-Dawley , Treatment Outcome
19.
Behav Brain Res ; 324: 41-50, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28212939

ABSTRACT

Cannabis sativa is one of the most widely recreational drugs and its use is more prevalent among depressed patients. Some studies reported that Cannabis has antidepressant effects while others showed increased depressive symptoms in Cannabis users. Therefore, the present study aims to investigate the effect of Cannabis extract on the depressive-like rats. Twenty four rats were divided into: control, rat model of depression induced by reserpine and depressive-like rats treated with Cannabis sativa extract (10mg/kg expressed as Δ9-tetrahydrocannabinol). The depressive-like rats showed a severe decrease in motor activity as assessed by open field test (OFT). This was accompanied by a decrease in monoamine levels and a significant increase in acetylcholinesterase activity in the cortex and hippocampus. Na+,K+-ATPase activity increased in the cortex and decreased in the hippocampus of rat model. In addition, a state of oxidative stress was evident in the two brain regions. This was indicated from the significant increase in the levels of lipid peroxidation and nitric oxide. No signs of improvement were observed in the behavioral and neurochemical analyses in the depressive-like rats treated with Cannabis extract. Furthermore, Cannabis extract exacerbated the lipid peroxidation in the cortex and hippocampus. According to the present findings, it could be concluded that Cannabis sativa aggravates the motor deficits and neurochemical changes induced in the cortex and hippocampus of rat model of depression. Therefore, the obtained results could explain the reported increase in the depressive symptoms and memory impairment among Cannabis users.


Subject(s)
Cannabis/adverse effects , Depression/chemically induced , Dronabinol/administration & dosage , Acetylcholinesterase/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Depression/metabolism , Disease Models, Animal , Dopamine/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Motor Activity , Norepinephrine/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Reserpine , Serotonin/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
20.
Asian Pac J Trop Med ; 10(5): 478-490, 2017 May.
Article in English | MEDLINE | ID: mdl-28647186

ABSTRACT

OBJECTIVE: To investigate the effect of two extracts of Bougainvillea spectabilis (B. spectabilis) flowers with yellow and pink/purple on brain oxidative stress and neuronal damage caused in rats by systemic rotenone injection. METHODS: Rotenone 1.5 mg/kg was given three times per week alone or in combination with B. spectabilis flowers extracts (25 mg or 50 mg) via the subcutaneous route for 2 weeks. Brain concentrations of the lipid peroxidation marker malondialdehyde (MDA), reduced glutathione, nitric oxide (nitrite), the pro-inflammatory cytokine interleukin-1beta (Il-1ß) as well as butyrylcholinesterase, and paraoxonase-1 (PON-1) activities, were determined. Histopathology and caspase-3 immunohistochemistry were also performed. RESULTS: Rotenone resulted in significant increases of brain MDA (the product of lipid peroxidation), and nitric oxide content along with decreased brain reduced glutathione. There were also marked and significant inhibition of brain PON-1 and BChE activities and increased Il-1ß in brain of rotenone-treated rats. B. spectabilis flowers extract itself resulted in brain oxidative stress increasing both lipid peroxidation and nitrite content whilst inhibiting PON-1 activity. The yellow flowers extract inhibited BChE activity and increased brain Il-1ß. When given to rotenone-treated rats, B. spectabilis extracts, however, decreased lipid peroxidation while their low administered doses increased brain GSH. Brain nitrite decreased by the pink extract but showed further increase by the yellow extract. Either extract, however, caused further inhibition of PON-1 activity while the yellow extract resulted in further inhibition of BChE activity. Histopathological studies indicated that both extracts protected against brain, liver and kidney damage caused by the toxicant. CONCLUSIONS: These data indicate that B. spectabilis flowers extracts exert protective effect against the toxic effects of rotenone on brain, liver and kidney. B. spectabilis flowers extracts decreased brain lipid peroxidation and prevented neuronal death due to rotenone and might thus prove the value in treatment of Parkinson's disease.

SELECTION OF CITATIONS
SEARCH DETAIL