Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Dose Response ; 21(1): 15593258231155787, 2023.
Article in English | MEDLINE | ID: mdl-36756150

ABSTRACT

Ulcerative colitis (UC) is an inflammatory bowel disease involving chronic and recurring colon inflammation. Current management protocols are limited by adverse effects or short-term symptomatic relief. We aimed to investigate the possible therapeutic prospect of low dose gamma (γ) irradiation or apigenin treatment in acetic acid-induced UC in rats. Induction of UC was carried out by installation of acetic acid intra-rectally. One hour post-induction, rats received a sole dose of γ-radiation (0.5 Gray) or were treated with apigenin (3 mg/kg/day, peroral) for 7 successive days. Antioxidant and anti-inflammatory effects of both agents were assessed via determination of colon malondialdehyde (MDA), reduced glutathione (GSH), total nitrate/nitrite (NOx), mucosal addressin cell adhesion molecule-1 (MAdCAM-1), and interleukin-1beta (IL-1ß) contents as well as myeloperoxidase (MPO) activity. Body weight (BW), colon weight/length (W/L) ratio, disease activity index (DAI), and histopathological changes were evaluated. Gamma irradiation and apigenin significantly ameliorated the acetic acid-induced biochemical and histopathological changes. Both therapeutic approaches significantly restored colon contents of the investigated biomarkers. They modulated BW, colon W/L ratio and DAI. This study proposes low dose γ-irradiation as a new therapeutic candidate for the management of UC. We also concluded that apigenin exhibited therapeutic benefits in UC management.

2.
Int Immunopharmacol ; 114: 109571, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36527875

ABSTRACT

Neuroinflammatory status produced via activation of toll like receptor-4 (TLR-4) and interleukin-17 receptor (IL-17R) is one of the principal mechanisms involved in dopaminergic neuronal loss in Parkinson's disease (PD). Activation of TLR-4 and IL-17R stimulates reactive oxygen species (ROS) and proinflammatory cytokines (IL-17, IL-1ß, TNFα, IL-6) production that augments neurodegeneration and reduces neuro-survival axis (TrKB/Akt/CREB/BDNF). So, reducing IL-17-driven neuroinflammation via secukinumab, monoclonal antibody against IL-17A, may be one of therapeutic approach for PD. Moreover, the aim was extended to delineate the possible neuroprotective mechanism involved against neuronal loss in rotenone induced PD in rats. Rats received 11 subcutaneous injection of rotenone (1.5 mg/kg) every other day for 21 consecutive days and treated with 2 subcutaneous injections of secukinumab (15 mg/kg) on day 9 and 15, one hour after rotenone administration. Treatment with secukinumab improved motor impairment and muscle incoordination induced by rotenone, as verified by open field and rotarod tests. Moreover, secukinumab attenuated neuronal loss and improve histopathological profile. Noteworthy, secukinumab reduces neuro-inflammatory status by hindering the interaction between IL and 17A and IL-17RA together with inhibiting the activation of TLR-4 and its downstream cascade including pS536-NFκB p65, IL-1ß and HMGB-1. Additionally, secukinumab stimulated neuro-survival signalling cascade via activation pY515-TrKB receptor and triggered upsurge in its downstream targets (pS473-Akt/pS133-CREB/BDNF). Furthermore, secukinumab increased striatal tyrosine hydroxylase immunoexpression, the rate limiting step in dopamine biosynthesis, to guard against dopaminergic neuronal loss. In conclusion, secukinumab exerts a neuroprotective effect against rotenone induced neuronal loss via inhibition IL17A/IL17RA interaction and HMGB-1/TLR-4/NF-κBp65/IL1ß signalling cascade, together with activation of TrKB/ Akt/CREB/BDNF axis.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Rats , Animals , Parkinson Disease/pathology , Rotenone , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Interleukin-17 , Brain-Derived Neurotrophic Factor , Toll-Like Receptor 4 , Proto-Oncogene Proteins c-akt , Receptor, trkB , Rats, Wistar , HMGB Proteins
3.
J Biochem Mol Toxicol ; 34(4): e22451, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31975531

ABSTRACT

Dapagliflozin (DAPA) is used for treating type 2 diabetes, whereas lansoprazole (LPZ) is used as a traditional antiulcer drug. The present study investigated the possible antidiabetic effects of LPZ on fortified diet-fed streptozotocin (FDF/STZ)-induced insulin-resistant diabetic rats. On the basis of the current results, it can be concluded that LPZ could be used as an add-on drug along with the conventional treatment for T2D as it showed beneficial effects in the current experimental model of insulin resistance.


Subject(s)
Benzhydryl Compounds/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Glucosides/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Lansoprazole/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Body Weight/drug effects , Cholesterol/blood , Cholesterol/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Food, Fortified/adverse effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , PPAR gamma/blood , Pancreas/metabolism , Pancreas/pathology , Random Allocation , Rats , Rats, Wistar
4.
Can J Physiol Pharmacol ; 98(4): 183-193, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31665614

ABSTRACT

Doxorubicin (DOX), an anthracycline antibiotic, is an important antineoplastic agent due to its high antitumor efficacy in hematological as well as in solid malignancies. The clinical use of DOX is limited due to its cardiotoxic effects. The present study aimed to investigate the possible protective effect of olmesartan (Olm), l-carnitine (L-CA), and their combination in cardiotoxicity induced by DOX in rats. Male albino rats were randomly divided into seven experimental groups (n = 8): group I: normal control, group II: L-CA, group III: Olm, group IV: DOX. The other three groups were treated with Olm (10 mg/kg), L-CA (300 mg/kg), and their combination for 2 weeks after induction of cardiotoxicity by a single dose of DOX (20 mg/kg). In the results, DOX showed a significant elevation in serum troponin I, creatine kinase-MB (CK-MB), and lactate dehydrogenase (LDH) together with increased inflammation manifested by the rise of tumor necrosis factor-alpha (TNF-α), intercellular adhesion molecules-1 (ICAM-1), interleukin IL-1ß (IL-1ß), myeloperoxidase (MPO), nuclear factor-kappa B (NF-κB), and transforming growth factor beta (TGF-ß) in cardiac tissues as well as DOX-induced oxidative stress by increasing in malondialdehyde (MDA) and decreasing in superoxide dismutase (SOD) and glutathione (GSH) in heart tissues. In addition, caspase-3 activity was boosted as indication of increased apoptosis. On the other hand, administration of L-CA and Olm attenuated the DOX-evoked disturbances in the abovementioned parameters. In addition, DOX exhibited echocardiographic changes and severe histopathological changes, which were significantly reversed by L-CA and Olm treatment. In conclusion, the present study data confirm the protective role of L-CA and Olm in DOX-induced cardiotoxicity, which may be related to its antioxidant, antiinflammatory, and antiapoptotic agents.


Subject(s)
Cardiotoxicity/drug therapy , Carnitine/pharmacology , Doxorubicin/pharmacology , Imidazoles/pharmacology , Protective Agents/pharmacology , Tetrazoles/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Creatine Kinase, MB Form/metabolism , Heart/drug effects , Inflammation/drug therapy , Inflammation/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Malondialdehyde/metabolism , Myocardium/metabolism , Oxidative Stress/drug effects , Rats , Troponin I/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Biochem Pharmacol ; 171: 113681, 2020 01.
Article in English | MEDLINE | ID: mdl-31669235

ABSTRACT

In the current investigation, a Parkinson's disease (PD) model was established by a single direct right intrastriatal injection of the 6-hydroxydopamine (OHDA) in male Wistar rats followed by 7 daily unilateral injection of angiotensin (Ang) 1-7 in the striatum. To confirm the putative role of Mas receptor (MasR), the selective antagonist A779 was also injected intrastriatally prior to Ang 1-7 injections and a correlation analysis was performed between MasR expression and the assessed parameters. Ang 1-7 upregulated MasR expression to correlate strongly with the improved rotarod (r = 0.95, p = 0.003) and spontaneous activity task (r = 0.99, p < 0.0001). This correlation extends to involve other effects of Ang 1-7, such as the increased striatal dopamine content (r = 0.98, p = 0.0005), substantia nigra pars compacta tyrosine hydroxylase immune-reactivity (r = 0.97, p = 0.001), active pY705-STAT3 (r = 0.99, p < 0.0001) and SOCS3 (r = 0.99, p < 0.0001). Conversely, Ang 1-7 inhibited inflammatory markers to correlate negatively with NF-κBp65 (r = -0.99, p < 0.0003) and its downstream targets, high mobility group box-1 (HMGB-1; r = -0.97, p = 0.002), receptor for advanced glycation end products (RAGE; r = -0.98, p = 0.0004), and TNF-α (r = -0.99, p < 0.0003), besides poly-ADP-ribose polymerase-1 (r = -0.99, p = 0.0002). In confirmation, the pre-administration of the selective MasR antagonist, A779, partially attenuated Ang 1-7-induced alterations towards 6-OHDA neurodegeneration. Collectively, our findings support a novel role for the anti-inflammatory capacity of the MasR axis to prove potential therapeutic relevance in PD via the upregulation/activation of MasR-dependent STAT3/SOCS3 cascade to negatively control the HMGB-1/RAGE/NF-κB axis hindering PD associated neuro-inflammation along with DA depletion and motor deficits.


Subject(s)
Angiotensin I/administration & dosage , Motor Activity/drug effects , Parkinsonian Disorders/metabolism , Peptide Fragments/administration & dosage , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Gene Expression/drug effects , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Male , NF-kappa B/metabolism , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Rats, Wistar , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Receptors, G-Protein-Coupled/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vasodilator Agents/administration & dosage
6.
Eur J Pharmacol ; 861: 172620, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31437429

ABSTRACT

The reno-protective effects of antidiabetic dipeptidyl peptidase (DPP)-4 inhibitors have been studied regarding their antioxidant and anti-inflammatory properties. However, the potential ability of saxagliptin to ameliorate renal injury by enhancing neovascularization has not been elucidated. To address this issue, saxagliptin (10 and 30 mg/kg) was administered to Wistar rats after the induction of renal ischaemia/reperfusion (I/R). Our results showed that saxagliptin operated through different axes to ameliorate I/R injury. By inhibiting DPP-4, saxagliptin maintained stromal cell-derived factor-1α expression and upregulated its chemokine receptor CXCR4 to trigger vasculogenesis through the enhanced migration of endothelial progenitor cells (EPCs). Additionally, this compound rescued the levels of glucagon-like peptide-1 and its downstream mediator cAMP to increase vascular endothelial growth factor (VEGF) and CXCR4 levels. Moreover, saxagliptin stimulated atrial natriuretic peptide/endothelial nitric oxide synthase to increase nitric oxide levels and provoke angiogenesis and renal vasodilation. In addition to inhibiting DPP-4, saxagliptin increased the renal kidney injury molecule-1/pY705-STAT3/hypoxia-inducible factor-1α/VEGF pathway to enhance angiogenesis. Similar to other gliptins, saxagliptin exerted its anti-inflammatory and antioxidant effects by suppressing the renal contents of p (S536)-nuclear factor-κB p65, tumour necrosis factor-α, monocyte chemoattractant protein-1, myeloperoxidase, and malondialdehyde while boosting the glutathione content. These events improved the histological structure and function of the kidney, as evidenced by decreased serum creatinine, blood urea nitrogen, and cystatin C and increased serum albumin. Accordingly, in addition to its anti-inflammatory and antioxidant activities, saxagliptin dose-dependently ameliorated I/R-induced renal damage by enhancing neovascularization through improved tissue perfusion and homing of bone marrow-derived EPCs to mediate repair processes.


Subject(s)
Adamantane/analogs & derivatives , Dipeptides/pharmacology , Kidney/blood supply , Kidney/drug effects , Neovascularization, Physiologic/drug effects , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Atrial Natriuretic Factor/metabolism , Cell Adhesion Molecules/metabolism , Chemokine CXCL12/metabolism , Cyclic AMP/metabolism , Dipeptides/therapeutic use , Disease Models, Animal , Glucagon-Like Peptide 1/metabolism , Glutathione/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Kidney/physiopathology , Male , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Rats , Rats, Wistar , Receptors, CXCR4/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
JGH Open ; 3(4): 281-289, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31406920

ABSTRACT

BACKGROUND: Inflammatory bowel diseases are immunologically mediated disorders of gastrointestinal tract, characterized by dysregulated immune responses that result in a chronic intestinal inflammation. The antiplatelet cilostazol (CS), a phosphodiesterase-III inhibitor, exerted a beneficial effect on several models of gastrointestinal diseases; however, the full mechanism of action in this context has not been unveiled. AIM: The current study aimed to elucidate the potential role of CS in a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis model. METHODS: Male Wistar rats were divided into a sham group and groups treated with sulfasalazine (500 mg/kg), CS (50 and 100 mg/kg), and a combination (sulfasalazine/CS 50 mg/kg). All treatments were administered orally 15 days, with TNBS rectal administration on the 11th day. RESULTS: TNBS-produced colitis manifested as a decrease in the epithelial junctional adhesion molecule-A (JAM-A) and as an increase in trefoil factor-3, ulcerative area, and colon mass index, parameters that collaborate with the gross macroscopic changes in colon tissue. In addition, TNBS increased hemeoxygenase-1, nuclear factor-kappa B, P-selectin, and myeloperoxidase, as well as the apoptotic ratio of Bax/Bcl-2. Administration of CS alone, especially at the high dose level, attenuated the severity of TNBS-induced colitis in a sulfasalazine-comparable manner. In addition, a better effect was mediated by the combination regimen, which succeeded in normalizing most of the measured parameters. CONCLUSION: CS protected the colon against TNBS through its anti-inflammatory and antiapoptotic effects along with maintaining cellular tight junctions (TJs). Furthermore, CS can be beneficial as an add-on drug with the conventional treatments of colitis.

8.
Clin Exp Pharmacol Physiol ; 46(12): 1141-1150, 2019 12.
Article in English | MEDLINE | ID: mdl-31408200

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) and nuclear receptor related-1 (Nurr1) play pivotal roles in the development and survival of dopaminergic neurons, and deficiencies in these genes may be involved in Parkinson's disease (PD) pathogenesis. Recently, anthelminthic benzimidazoles were shown to promote HIF-1α transcription in vitro and were proposed to activate Nurr1 via their benzimidazole group. Therefore, the aim of this study was to explore the neuroprotective effects of albendazole (ABZ), an anthelminthic benzimidazole, in a rotenone model of Parkinson's disease (PD). Rotenone (1.5 mg/kg) was subcutaneously injected into rats every other day for a period of 21 days, resulting in the development of the essential features of PD. In addition to rotenone, ABZ (10 mg/kg) was administered orally starting from the 11th day. Treatment of rats with ABZ markedly mitigated rotenone-induced histological alterations in substantia nigra (SN), restored striatal dopamine (DA) level and motor functions and decreased the expression of α-synuclein (a disease marker protein). ABZ also enhanced expression of Hypoxia-inducible factor-1 alpha (HIF-1α) in the SN along with its downstream target, vascular endothelial growth factor, promoting neuronal survival. Similarly, ABZ augmented nuclear receptor related-1 (Nurr1) expression in the SN and increased transcriptional activation of Nurr1-controlled genes, which are essential for regulation of DA synthesis; additionally, expression of neurotoxic proinflammatory cytokines that induce neuronal death was suppressed. In conclusion, the present study suggests that ABZ exerts a neuroprotective effect in a rotenone-induced PD model associated with HIF-1α and Nurr1 activation and thus may be a viable candidate for treating PD.


Subject(s)
Albendazole/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neuroprotection/drug effects , Neuroprotection/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinson Disease , Albendazole/therapeutic use , Animals , Behavior, Animal/drug effects , Cell Death/drug effects , Cell Death/genetics , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Molecular Targeted Therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Rats , Rats, Wistar , Rotenone
9.
Neurotox Res ; 35(3): 635-653, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30673988

ABSTRACT

L-dopa is still considered as the gold standard therapy for Parkinson's disease (PD); however, L-dopa-induced dyskinesia (LID) is a serious complication of long-term L-dopa treatment. The present study investigated the therapeutic potential of sitagliptin and liraglutide in comparisons with L-dopa against PD. In addition, their capacity to modulate L-dopa dose and/or side effects was investigated, too. Rats were injected with rotenone (3 mg/kg/day, s.c.) for 10 consecutive days to induce the experimental PD. The rotenone-treated rats were administered sitagliptin (30 mg/kg/day, p.o.) and liraglutide (50 µg/kg, s.c.) for 16 days either alone or together with L-dopa/carbidopa (50/25 mg/kg/day, i.p.). Scoring of LID was done on days 2, 4, 8, 12, and 16 in all L-dopa-treated groups. Twenty-four hours after the last administered dose of tested drugs, the behavior of rats in each group was screened by using the open-field test. Sitagliptin and liraglutide revealed marked attenuation of LID scores; in addition, they markedly increased animals' motor performance. Moreover, they preserved substantia nigra pars compacta (SNpc) tyrosine hydroxylase (TH) and vesicular monoamine transporter 2-positive (VMAT2) cells with prominent increase of the striatal dopamine (DA) content. On the other hand, they significantly decreased nigral neuromelanin (NM)-positive cells, activated microglia, gliosis, and other pathological changes. In conclusion, sitagliptin and liraglutide could be a promising therapeutic challenger in PD, modifying L-dopa effect and/or allowing the use of L-dopa with fewer side effects.


Subject(s)
Antiparkinson Agents/pharmacology , Carbidopa/pharmacology , Dyskinesia, Drug-Induced/prevention & control , Levodopa/pharmacology , Liraglutide/pharmacology , Parkinsonian Disorders/drug therapy , Sitagliptin Phosphate/pharmacology , Animals , Antiparkinson Agents/adverse effects , Carbidopa/adverse effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Drug Combinations , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/pathology , Levodopa/adverse effects , Liraglutide/adverse effects , Male , Motor Activity/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , Random Allocation , Rats, Wistar , Rotenone , Sitagliptin Phosphate/adverse effects
10.
PLoS One ; 13(9): e0203837, 2018.
Article in English | MEDLINE | ID: mdl-30260985

ABSTRACT

Countless neurodegenerative diseases are associated with perverse multiple targets of cyclic nucleotide signalling, hastening neuronal death. Cilostazol, a phosphodiesterase-III inhibitor, exerts neuroprotective effects against sundry models of neurotoxicity, however, its role against Huntington's disease (HD) has not yet been tackled. Hence, its modulatory effect on several signalling pathways using the 3-nitropropionic acid (3-NP) model was conducted. Animals were injected with 3-NP (10 mg/kg/day, i.p) for two successive weeks with or without the administration of cilostazol (100 mg/kg/day, p.o.). Contrary to the 3-NP effects, cilostazol largely preserved striatal dopaminergic neurons, improved motor coordination, and enhanced the immunohistochemical reaction of tyrosine hydroxylase enzyme. The anti-inflammatory effect of cilostazol was documented by the pronounced reduction of the toll like receptor-4 (TLR-4) protein expression and the inflammatory cytokine IL-6, but with a marked elevation in IL-10 striatal contents. As a consequence, cilostazol reduced IL-6 downstream signal, where it promoted the level of suppressor of cytokine signalling 3 (SOCS3), while abated the phosphorylation of Janus Kinase 2 (JAK-2) and Signal transducers and activators of transcription 3 (STAT-3). Phosphorylation of the protein kinase B/glycogen synthase kinase-3ß/cAMP response element binding protein (Akt/GSK-3ß/CREB) cue is another signalling pathway that was modulated by cilostazol to further signify its anti-inflammatory and antiapoptotic capacities. The latter was associated with a reduction in the caspase-3 expression assessed by immunohistochemical assay. In conclusion the present study provided a new insight into the possible mechanisms by which cilostazol possesses neuroprotective properties. These intersecting mechanisms involve the interference between TLR-4, IL-6-IL-10/JAK-2/STAT-3/SOCS-3, and Akt/GSK-3ß/CREB signalling pathways.


Subject(s)
Cilostazol/pharmacology , Huntington Disease/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , CREB-Binding Protein/drug effects , CREB-Binding Protein/metabolism , Cilostazol/therapeutic use , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Huntington Disease/metabolism , Interleukin-6/metabolism , Janus Kinase 2/drug effects , Janus Kinase 2/metabolism , Male , Neuroprotective Agents/pharmacology , Nitro Compounds/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Propionates/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , STAT2 Transcription Factor/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/drug effects , Suppressor of Cytokine Signaling 3 Protein/metabolism , Toll-Like Receptor 4/metabolism
11.
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
12.
Biochem Pharmacol ; 151: 126-134, 2018 05.
Article in English | MEDLINE | ID: mdl-29428223

ABSTRACT

MAS receptor (MASR), expressed in several brain areas, conferred neuroprotection against neurodegenerative disorders when activated by angiotensin (Ang) 1-7; however, its role in Parkinson's disease (PD) remains elusive. Intra-striatal post-administration of Ang1-7, using a 6-hydroxydopamine (OHDA) PD model, improved motor performance and muscle coordination. On the molecular level, Ang1-7 upregulated the striatal expression of MASR and caused upsurge in its downstream targets (p-PI3K/p-Akt/p-CREB/BDNF) to phosphorylate TrKB, which in a positive feedback upregulates MASR. Moreover, Ang1-7 increased substantia nigral tyrosine hydroxylase (TH) expression and striatal dopamine (DA) content to indicate the preservation of the dopaminergic neuronal signal. This effect extended to inhibit the striatal expression of Ang II type-1 receptor (AT-1R) to hold the neurodegenerative effect and to boost Ang1-7 anti-inflammatory/antioxidant effects by abating NADPH oxidase, along with lipid peroxidation. Indeed, Ang1-7 was able to decrease p-MAPK p38/NF-κB p65 to level the inflammatory and oxidative stress events off. The Ang1-7-mediated activation of MASR cue and the suppression of the AT-1R cascade were partially reversed by the intrastartial injection of A-779, a MASR antagonist. The current data suggests a novel therapeutic potential for the Ang1-7 against neurotoxicity associated motor impairment related to PD. The anti-parkinsonian effect of Ang1-7, is in part, mediated by its binding to MASR and the initiation of PI3K/Akt/CREB/BDNF/TrKB cue to increase DA synthesis, besides the downregulation/inhibition of AT-1R/MAPK p38/NF-κB p65/NADPH oxidase pathway.


Subject(s)
Angiotensin I/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , NF-kappa B/metabolism , Parkinsonian Disorders/drug therapy , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Male , Oxidopamine , Parkinsonian Disorders/metabolism , Proto-Oncogene Mas , Rats, Wistar , Signal Transduction/drug effects
13.
Mol Neurobiol ; 55(5): 4078-4089, 2018 May.
Article in English | MEDLINE | ID: mdl-28585189

ABSTRACT

Recently, it has been shown that both decreased nuclear receptor-related 1 (Nurr1) expression and thrombin accumulation are involved in the degeneration of dopaminergic neurons in Parkinson's disease (PD). The new anticoagulant dabigatran etexilate (DE) is a direct thrombin inhibitor that owns benzimidazole group, which has been proposed to activate Nurr1. In the present study, we examined the neuroprotective effects of DE in rotenone model of PD. Rotenone was injected subcutaneously at a dose of 1.5 mg/kg every other day for 21 days. An oral regimen of DE (15 mg/kg) was started after the 5th rotenone injection following the manifestations of PD. Treatment of PD rats with DE mitigated rotenone-induced neuronal degeneration and restored striatal dopamine level with motor recovery. As well, DE enhanced Nurr1 expression in substantia nigra along with increasing transcriptional activation of Nurr1-controlled genes namely tyrosine hydroxylase, vascular monoamine transporter, glial cell line-derived neurotrophic factor, and its receptor gene c-Ret, which are critical for development and maintenance of dopaminergic neurons. DE also suppressed thrombin accumulation in substantia nigra. Both effects probably contributed to repressing neurotoxic proinflammatory cytokines, which was manifested by decreased level of nuclear factor kappa beta and tumor necrosis factor alpha. In conclusion, the present results suggest that DE could possess significant neuroprotective and regenerative effects in a rotenone-induced PD animal model as consequence of Nurr1 activation and thrombin inhibition.


Subject(s)
Dabigatran/pharmacology , Neuroprotective Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinson Disease/metabolism , Thrombin/pharmacology , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Inflammation/pathology , Male , Neostriatum/metabolism , Neostriatum/pathology , Parkinson Disease/pathology , Rats, Wistar , Rotenone , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
14.
Front Pharmacol ; 8: 735, 2017.
Article in English | MEDLINE | ID: mdl-29081748

ABSTRACT

Statins were reported to lower the Coenzyme Q10 (CoQ10) content upon their inhibition of HMG-CoA reductase enzyme and both are known to possess neuroprotective potentials; therefore, the aim is to assess the possible use of CoQ10 as an adds-on therapy to rosuvastatin to improve its effect using global I/R model. Rats were allocated into sham, I/R, rosuvastatin (10 mg/kg), CoQ10 (10 mg/kg) and their combination. Drugs were administered orally for 7 days before I/R. Pretreatment with rosuvastatin and/or CoQ10 inhibited the hippocampal content of malondialdehyde, nitric oxide, and boosted glutathione and superoxide dismutase. They also opposed the upregulation of gp91phox, and p47phox subunits of NADPH oxidase. Meanwhile, both agents reduced content/expression of TNF-α, iNOS, NF-κBp65, ICAM-1, and MPO. Besides, all regimens abated cytochrome c, caspase-3 and Bax, but increased Bcl-2 in favor of cell survival. On the molecular level, they increased p-Akt and its downstream target p-FOXO3A, with the inhibition of the nuclear content of FOXO3A to downregulate the expression of Bim, a pro-apoptotic gene. Additionally, both treatments downregulate the JNK3/c-Jun signaling pathway. The effect of the combination regimen overrides that of either treatment alone. These effects were reflected on the alleviation of the hippocampal damage in CA1 region inflicted by I/R. Together, these findings accentuate the neuroprotective potentials of both treatments against global I/R by virtue of their rigorous multi-pronged actions, including suppression of hippocampal oxidative stress, inflammation, and apoptosis with the involvement of the Akt/FOXO3A/Bim and JNK3/c-Jun/Bax signaling pathways. The study also nominates CoQ10 as an adds-on therapy with statins.

15.
Inflammopharmacology ; 25(3): 369-382, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28258522

ABSTRACT

The present study investigated the possible relationship between pro-inflammatory cytokines and programmed nigral neuronal death in rotenone model of Parkinson's disease (PD). Sitagliptin and liraglutide efficacy to inhibit the inflammatory-apoptotic degenerative process were investigated, too. The experimental PD were induced in male albino rats by ten subcutaneously injections of rotenone (3 mg/kg/day, s.c). All treatment drugs were administered for 16 days after induction of Parkinson rat's model. Sitagliptin and liraglutide were administered in three different dose levels (10-20-30 mg/kg, p.o), (25-50-100 µg/kg, s.c), respectively. Cylindrical and catalepsy tests were used to detect the optimum dose response of each drug. Sitagliptin (30 mg/kg/day, p.o) and liraglutide (50 µg/kg, s.c.) showed statistically significant (p ≤ 0.05) effect on behavioral activity. Where both doses improved the motor performance significantly in comparison with other doses in both cylindrical and catalepsy tests. Furthermore, they reversed rotenone-induced nigral neuronal loss, associated with marked decrease of pro-inflammatory cytokines: interleukin (IL)-1ß, IL-6, transforming growth factor (TGF)-ß1, together with a significant increase of striatal dopamine, nigral glial cell line-derived neurotrophic factor (GDNF), and tyrosine hydroxylase positive (TH+) cells. Moreover, the pro-apoptotic environment in nigrostriatal tissues was abrogated significantly, as the pro-apoptotic protein Bax decreased along with the anti-apoptotic protein Bcl-2 increased. In conclusion, sitagliptin and liraglutide represent a promising strategy to mitigate the progression of PD by their anti-inflammatory, anti-apoptotic neurotrophic and neurogenic mechanistic activities.


Subject(s)
Brain/drug effects , Liraglutide/pharmacology , Parkinson Disease/drug therapy , Rotenone/pharmacology , Sitagliptin Phosphate/pharmacology , Animals , Apoptosis/drug effects , Brain/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Parkinson Disease/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rodentia , Transforming Growth Factor beta1/metabolism , Tyrosine 3-Monooxygenase/metabolism , bcl-2-Associated X Protein/metabolism
16.
Int J Radiat Biol ; 93(5): 553-560, 2017 05.
Article in English | MEDLINE | ID: mdl-28067097

ABSTRACT

OBJECTIVE: Fibromyalgia is a prevalent disorder characterized by chronic widespread pain and complex symptoms. This study was conducted to investigate the potential therapeutic effect of low-dose irradiation (LDI) alone or in combination with duloxetine on the reserpine-induced fibromyalgia in rats. METHODS: Fibromyalgia was induced by administration of reserpine (1 mg/kg/s.c) for 3 consecutive days. Duloxetine (30 mg/kg, p.o) was administered 60 min before a forced swimming test (FST), and rats were exposed to a single dose of γ-radiation (0.5 Gy) 1 day before the FST. RESULTS: Reserpine significantly increased immobility time in the FST, decreased the amount of 5-hydroxytryptamine, dopamine, and norepinephrine in cerebral cortex. It also increased malondialdehyde and nitric oxide and reduced glutathione contents in brain tissue. LDI alone or combined with duloxetine completely antagonized reserpine-induced fibromyalgia as assessed by the measured parameters. One of the most significant findings in this study was that the therapeutic effect of duloxetine was more pronounced by its combination with LDI. A possible mechanism of action of LDI and duloxetine responsible for their therapeutic effect was discussed. CONCLUSION: On the basis of the presented evidences, it could be concluded that LDI alone or combined with duloxetine could be of value in the management of fibromyalgia.


Subject(s)
Chemoradiotherapy/methods , Duloxetine Hydrochloride/administration & dosage , Fibromyalgia/diagnosis , Fibromyalgia/therapy , Gamma Rays/therapeutic use , Analgesics/administration & dosage , Animals , Fibromyalgia/chemically induced , Male , Radiotherapy Dosage , Rats , Rats, Wistar , Reserpine , Treatment Outcome
17.
Can J Physiol Pharmacol ; 94(2): 131-139, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26695545

ABSTRACT

Cyclophosphamide (CP) is a cytotoxic alkylating agent used in the treatment of malignant diseases and autoimmune disorders. Its clinical use is limited to its marked cardiorenal toxicity. The present study aimed to investigate the possible protective role of taurine (Tau; 200 mg·kg-1 per day, i.p.) on CP-induced cardiorenal toxicity. CP (200 mg·kg-1) was administered as a single intraperitoneal injection whereas; Tau was administered for 3 weeks on a daily basis. The results showed that CP produced an elevation in serum activities of creatine kinase, creatine kinase isoenzyme, lactate dehydrogenase, creatinine as well as blood urea nitrogen. CP also induced an elevation in the oxidative stress markers viz. elevation in the serum lipid peroxides level (measured as malondialdehyde; MDA) and reduction in reduced glutathione level and superoxide dismutase activity in both heart and renal tissue. On the other hand, administration of Tau attenuated the CP-evoked disturbances in the above mentioned parameters. In addition, CP exhibited electrocardiographic (ECG) changes, which were significantly reversed by Tau treatment. Finally, the histopathological examination emphasized the obtained results. In conclusion, Tau is suggested to be a potential candidate to ameliorate CP-induced cardiorenal toxicity that may be related to its antioxidant activity.

18.
Eur J Pharmacol ; 723: 330-8, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24315930

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disorder that leads to disturbances of cognitive functions. Although the primary cause of AD remains unclear, brain acetylcholine deficiency, oxidative stress and neuroinflammation may be considered the principal pathogenic factors. The present study was constructed to investigate the anti-amnestic effect of pyrrolidine dithiocarbamate (PDTC) on scopolamine-induced behavioral, neurochemical and biochemical changes in rats. PDTC (50 and 100mg/kg) and donepezil (2.5mg/kg) were orally administered for 14 successive days. Dementia was induced at the end of the treatment period by a single injection of scopolamine (20mg/kg; i.p.), and Y-maze test was conducted 30min thereafter. Rats were then sacrificed and homogenates of cortical and hippocampal tissues were used for the estimation of noradrenaline, dopamine, serotonin and heat shock protein 70 contents along with acetylcholinesterase activity. In addition, certain oxidative stress markers, pro-inflammatory and anti-inflammatory cytokines were assessed. Histological examination of cortical and hippocampal tissues was also performed. Scopolamine resulted in memory impairment that was coupled by alterations in the estimated neurotransmitters, heat shock protein 70, acetylcholinesterase activity, oxidative stress as well as inflammatory biomarkers. Histological analysis revealed serious damaging effects of scopolamine on the structure of cerebral cortex and hippocampus. Pretreatment of rats with PDTC in both doses mitigated scopolamine-induced behavioral, biochemical, neurochemical and histological changes in a manner comparable to donepezil. The observed anti-amnestic effect of PDTC makes it a promising candidate for clinical trials in patients with cognitive impairment.


Subject(s)
Cognition Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Pyrrolidines/therapeutic use , Thiocarbamates/therapeutic use , Alzheimer Disease , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Dopamine/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Male , Neuroprotective Agents/pharmacology , Norepinephrine/metabolism , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Scopolamine , Serotonin/metabolism , Thiocarbamates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...