Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 16(9)2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37765022

ABSTRACT

Cadmium is an environmental toxicant that instigates cognitive deficits with excessive glutamate excitatory neuroactivity in the brain. Topiramate, a glutamate receptor antagonist, has displayed favorable neuroprotection against epilepsy, cerebral ischemia, and Huntington's disease; however, its effect on cadmium neurotoxicity remains to be investigated. In this study, topiramate was tested for its potential to combat the cognitive deficits induced by cadmium in rats with an emphasis on hippocampal oxidative insult, apoptosis, and autophagy. After topiramate intake (50 mg/kg/day; p.o.) for 8 weeks, behavioral disturbances and molecular changes in the hippocampal area were explored. Herein, Morris water maze, Y-maze, and novel object recognition test revealed that topiramate rescued cadmium-induced memory/learning deficits. Moreover, topiramate significantly lowered hippocampal histopathological damage scores. Mechanistically, topiramate significantly replenished hippocampal GLP-1 and dampened Aß42 and p-tau neurotoxic cues. Notably, it significantly diminished hippocampal glutamate content and enhanced acetylcholine and GABA neurotransmitters. The behavioral recovery was prompted by hippocampal suppression of the pro-oxidant events with notable activation of SIRT1/Nrf2/HO-1 axis. Moreover, topiramate inactivated GSK-3ß and dampened the hippocampal apoptotic changes. In tandem, stimulation of hippocampal pro-autophagy events, including Beclin 1 upregulation, was triggered by topiramate that also activated AMPK/mTOR pathway. Together, the pro-autophagic, antioxidant, and anti-apoptotic features of topiramate contributed to its neuroprotective properties in rats intoxicated with cadmium. Therefore, it may be useful to mitigate cadmium-induced cognitive deficits.

2.
Pharmaceuticals (Basel) ; 16(7)2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37513918

ABSTRACT

Cadmium (Cd) is a widespread environmental pollutant that triggers testicular dysfunction. Dapagliflozin is a selective sodium-glucose co-transporter-2 inhibitor with notable antioxidant and anti-apoptotic features. It has shown marked cardio-, reno-, hepato-, and neuroprotective effects. Yet, its effect on Cd-evoked testicular impairment has not been examined. Hence, the goal of the current study was to investigate the potential positive effect of dapagliflozin against Cd-induced testicular dysfunction in rats, with an emphasis on autophagy, apoptosis, and oxidative insult. Dapagliflozin (1 mg/kg/day) was given by oral gavage, and testicular dysfunction, impaired spermatogenesis, and biomolecular events were studied via immunohistochemistry, histopathology, and ELISA. The current findings demonstrated that dapagliflozin improved relative testicular weight, serum testosterone, and sperm count/motility and reduced sperm abnormalities, signifying mitigation of testicular impairment and spermatogenesis disruption. Moreover, dapagliflozin attenuated Cd-induced histological abnormalities and preserved testicular structure. The testicular function recovery was prompted by stimulating the cytoprotective SIRT1/Nrf2/HO-1 axis, lowering the testicular oxidative changes, and augmenting cellular antioxidants. As regards apoptosis, dapagliflozin counteracted the apoptotic machinery by downregulating the pro-apoptotic signals together with Bcl-2 upregulation. Meanwhile, dapagliflozin reactivated the impaired autophagy, as seen by a lowered accumulation of SQSTM-1/p62 and Beclin 1 upregulation. In the same context, the testicular AMPK/mTOR pathway was stimulated as evidenced by the increased p-AMPK (Ser487)/total AMPK ratio alongside the lowered p-mTOR (Ser2448)/total mTOR ratio. Together, the favorable mitigation of Cd-induced testicular impairment/disrupted spermatogenesis was driven by the antioxidant, anti-apoptotic, and pro-autophagic actions of dapagliflozin. Thus, it could serve as a tool for the management of Cd-evoked testicular dysfunction.

3.
Int J Nanomedicine ; 17: 3967-3987, 2022.
Article in English | MEDLINE | ID: mdl-36105617

ABSTRACT

Introduction: Rituximab (RTX) and recombinant human myelin basic protein (rhMBP) were proven to be effective in ameliorating the symptoms of multiple sclerosis (MS). In this study, a nanoformulation containing rhMBP with RTX on its surface (Nano-rhMBP-RTX) was prepared and investigated in comparison with other treatment groups to determine its potential neuro-protective effects on C57BL/6 mice after inducing experimental autoimmune encephalomyelitis (EAE). Methods: EAE was induced in the corresponding mice by injecting 100 µL of an emulsion containing complete Freund's adjuvant (CFA) and myelin oligodendrocyte glycoprotein (MOG). The subjects were weighed, scored and subjected to behavioural tests. After reaching a clinical score of 3, various treatments were given to corresponding EAE-induced and non-induced groups including rhMBP, RTX, empty nanoparticle prepared by poly (lactide-co-glycolide) (PLGA) or the prepared nanoformulation (Nano-rhMBP-RTX). At the end of the study, biochemical parameters were also determined as interferon-γ (IFN-γ), myeloperoxidase (MPO), interleukin-10 (IL-10), interleukin-4 (IL-4), tumor necrosis factor alpha (TNF-α), nuclear factor kappa B (NF-kB), brain derived neurotrophic factor (BDNF), 2', 3' cyclic nucleotide 3' phosphodiesterase (CNP) and transforming growth factor beta (TGF-ß) along with some histopathological analyses. Results: The results of the Nano-rhMBP-RTX group showed promising outcomes in terms of reducing the clinical scores, improving the behavioural responses associated with improved histopathological findings. Elevation in the levels of IL-4, CNP and TGF-ß was also noticed along with marked decline in the levels of NF-kB and TNF-α. Conclusion: Nano-rhMBP-RTX treated group ameliorated the adverse effects induced in the EAE model. The effectiveness of this formulation was demonstrated by the normalization of EAE-induced behavioral changes and aberrant levels of specific biochemical markers as well as reduced damage of hippocampal tissues and retaining higher levels of myelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Interleukin-4 , Mice , Mice, Inbred C57BL , Myelin Basic Protein , NF-kappa B , Rituximab/pharmacology , Rituximab/therapeutic use , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha/therapeutic use
4.
Mol Neurobiol ; 59(7): 4562-4577, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35578102

ABSTRACT

Postmenopausal hormone-related cognitive decline has gained an immense interest to explore the underlying mechanisms and potential therapies. The current work aimed to study the possible beneficial effect of rosuvastatin (ROS) on the cognitive decline induced by ovariectomy in rats. Four groups were used as follows: control group, control + rosuvastatin, ovariectomy, and ovariectomy + rosuvastatin. After sham operation or ovariectomy, rats were given saline or oral dosages of ROS (2 mg/kg) every day for 30 days. The cognitive functions were assessed using the Morris water maze paradigm, Y-maze test, and new object recognition test. After rat killing, TLR4, caspase-8, and NLRP mRNA expression and protein levels of ASC, AIM2, caspase-1, NLRP1, and PKR were measured in hippocampus. This was complemented by the estimation of tissue content of NF-κΒ, IL-1ß, and IL-18 and serum lipid profile quantification. Rosuvastatin showed a promising potential for halting the cognitive impairments induced by estrogen decline through interfering with the TLR4/NF-κΒ/NLRP1/3 axis and inflammasomes activation and the subsequent pyroptosis. This was complemented by the amendment in the deranged lipid profile. Rosuvastatin may exert a beneficial role in attenuating the inflammatory and apoptotic signaling mechanisms associated with postmenopausal cognitive decline. Further investigations are needed to unveil the relationship between deranged plasma lipids and cognitive function.


Subject(s)
Cognitive Dysfunction , Inflammasomes , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , DNA-Binding Proteins/metabolism , Female , Humans , Inflammasomes/metabolism , Lipids , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins , Ovariectomy , Rats , Reactive Oxygen Species , Rosuvastatin Calcium/pharmacology , Rosuvastatin Calcium/therapeutic use , Toll-Like Receptor 4/metabolism
5.
Front Pharmacol ; 12: 735165, 2021.
Article in English | MEDLINE | ID: mdl-34690772

ABSTRACT

Mitochondrial oxidative status exerts an important role in modulating glia-neuron interplay during epileptogenesis. Trimetazidine (TMZ), a well-known anti-ischemic drug, has shown promising potential against a wide range of neurodegenerative disorders including epilepsy. Nevertheless, the exact mechanistic rationale behind its anti-seizure potential has not been fully elucidated yet. Herein, the impact of TMZ against mitochondrial oxidative damage as well as glutamate homeostasis disruption in the hippocampus has been investigated in rats with lithium/pilocarpine (Li/PIL) seizures. Animals received 3 mEq/kg i.p. LiCl3 followed by PIL (single i.p.; 150 mg/kg) 20 h later for induction of seizures with or without TMZ pretreatment (25 mg/kg; i.p.) for five consecutive days. Seizure score and seizure latency were observed. Mitochondrial redox status as well as ATP and uncoupling protein 2 was recorded. Moreover, glutamate homeostasis was unveiled. The present findings demonstrate the TMZ-attenuated Li/PIL seizure score and latency. It improved mitochondrial redox status, preserved energy production mechanisms, and decreased reactive astrocytes evidenced as decreased glial fibrillary acidic protein immune-stained areas in hippocampal tissue. In addition, it modulated phosphorylated extracellular signal-regulated kinases (p-ERK1/2) and p-AMP-activated protein kinase (p-AMPK) signaling pathways to reflect a verified anti-apoptotic effect. Consequently, it upregulated mRNA expression of astroglial glutamate transporters and reduced the elevated glutamate level. The current study demonstrates that TMZ exhibits robust anti-seizure and neuroprotective potentials. These effects are associated with its ability to modulate mitochondrial redox status, boost p-ERK1/2 and p-AMPK signaling pathways, and restore glutamate homeostasis in hippocampus.

6.
J Biochem Mol Toxicol ; 35(7): e22796, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33942446

ABSTRACT

Eprosartan (EPRO), an angiotensin receptor type-1 (AT-1) blocker, exhibited neuroprotective activities in ischemic stroke resulting from focal cerebral ischemia in rats. The current study aimed to clarify the neuroprotective role of EPRO in middle carotid artery occlusion (MCAO)-induced ischemic stroke in rats. Fifty-six male Wistar rats were divided into four groups (n = 14 per group): sham-operated group, sham receiving EPRO (60 mg/kg/day, po) group, ischemia-reperfusion (IR) group, and IR receiving EPRO (60 mg/kg/day, po) group. MCAO led to a remarkable impairment in motor function together with stimulation of inflammatory and apoptotic pathways in the hippocampus of rats. After MCAO, the AT1 receptor in the brain was stimulated, resulting in activation of Janus kinase 2/signal transducers and activators of transcription 3 signaling generating more neuroinflammatory milieu and destructive actions on the hippocampus. Augmentation of caspase-3 level by MCAO enhanced neuronal apoptosis synchronized with neurodegenerative effects of oxidative stress biomarkers. Pretreatment with EPRO opposed motor impairment and decreased oxidative and apoptotic mediators in the hippocampus of rats. The anti-inflammatory activity of EPRO was revealed by downregulation of nuclear factor-kappa B and tumor necrosis factor-ß levels and (C-X-C motif) ligand 1 messenger RNA (mRNA) expression. Moreover, the study confirmed the role of EPRO against a unique pathway of hypoxia-inducible factor-1α and its subsequent inflammatory mediators. Furthermore, upregulation of caveolin-1 mRNA level was also observed along with decreased oxidative stress marker levels and brain edema. Therefore, EPRO showed neuroprotective effects in MCAO-induced cerebral ischemia in rats via attenuation of oxidative, apoptotic, and inflammatory pathways.


Subject(s)
Acrylates/pharmacology , Brain/metabolism , Cerebrovascular Disorders/prevention & control , Imidazoles/pharmacology , Neuroprotective Agents/pharmacology , Thiophenes/pharmacology , Animals , Brain/pathology , Cerebrovascular Disorders/metabolism , Cerebrovascular Disorders/pathology , Male , Nerve Tissue Proteins/metabolism , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects
7.
J Pharmacol Sci ; 146(3): 136-148, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34030796

ABSTRACT

Despite the documented renoprotective effect of pentoxifylline (PTX), a non-selective phosphodiesterase-4 inhibitor, the studies appraised only its anti-inflammatory/-oxidant/-apoptotic capacities without assessment of the possible involved trajectories. Here, we evaluated the potential role of galectin-3 and the ASK-1/NF-κB p65 signaling pathway with its upstream/downstream signals in an attempt to unveil part of the cascades involved in the renotherapeutic effect using a renal bilateral ischemia/reperfusion (I/R) model. Rats were randomized into sham-operated, renal I/R (45 min/72 h) and I/R + PTX (100 mg/kg; p.o). Post-treatment with PTX improved renal function and abated serum levels of cystatin C, creatinine, BUN and renal KIM-1 content, effects that were reflected on an improvement of the I/R-induced renal histological changes. On the molecular level, PTX reduced renal contents of galectin-3, ASK-1 with its downstream molecule JNK and ERK1/2, as well as NF-κB p65 and HMGB1. This inhibitory effect extended also to suppress neutrophil infiltration, evidenced by diminishing ICAM-1 and MPO, as well as inflammatory cytokines (TNF-α/IL-18), oxidative stress (MDA/TAC), and caspase-3. The PTX novel renotherapeutic effect involved in part the inhibition of galectin-3 and ASK-1/JNK and ERK1/2/NF-κB/HMGB-1 trajectories to mitigate renal I/R injury and to provide basis for its anti-inflammatory, antioxidant, and anti-apoptotic impacts.


Subject(s)
Galectin 3/metabolism , HMGB1 Protein/metabolism , Kidney/blood supply , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor RelA/metabolism , Animals , Anti-Inflammatory Agents , Antioxidants , Disease Models, Animal , Male , Rats, Wistar
8.
Eur J Pharmacol ; 895: 173872, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33465355

ABSTRACT

In the present investigation, we tested the hypothesis that suppression of the phospho-extracellular signal regulated kinase (pERK1/2)-nuclear factor kappa (NFκ)-B signaling, subsequent to tumor necrosis factor-α (TNF-α) inhibition, underlies thalidomide (TLM) mediated neuroprotection. Male Wistar rats (250-280 g) were divided into five groups: (1) sham; (2) negative control receiving TLM (5µg/1µl/site) and 3 groups of ischemia-reperfusion (IR) injury rats pretreated with: (3) vehicle (DMSO 100%); (4) TLM (5µg/1µl/site) or (5) PD98059 (0.16µg/1µl/site). IR rats were subjected to occlusion of both common carotid arteries for 45 min followed by reperfusion for 24 h. Drugs and/or vehicles were administered by unilateral intrahippocampal injection after removal of the carotid occlusion and at the beginning of the reperfusion period. IR rats exhibited significant infarct size, histopathological damage, memory impairment, motor incoordination and hyperactivity. Unilateral intra-hippocampal TLM ameliorated these behavioral deficits along with the following ex vivo hippocampal effects: (i) abrogation of the IR-evoked elevations in hippocampal TNF-α, pERK1/2, NFκB, BDNF, iNOS contents and (ii) partial restoration of the reduced anti-inflammatory cytokine IL-10 and p-nNOS S852. These neurochemical effects, which were replicated by the pERK1/2 inhibitor PD98059, likely underlie the reductions in c-Fos and caspase-3 levels as well as the anti-apoptotic effect of TLM in the IR model. These results suggest a crucial anti-inflammatory role for pERK1/2 inhibition in the salutary neuronal and behavioral effects of TLM in a model of brain IR injury.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Ischemia/drug therapy , Hippocampus/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Thalidomide/pharmacology , Animals , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Male , Memory/drug effects , Motor Activity/drug effects , Phosphorylation , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
9.
Chem Biol Interact ; 335: 109368, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33412153

ABSTRACT

Dapagliflozin, a selective sodium-glucose co-transporter 2 (SGLT2) inhibitor, has featured marked anti-inflammatory effects in murine models of myocardial infarction, renal injury, and neuroinflammation. Yet, its potential impact on the pathogenesis of inflammatory bowel diseases (IBD) has not been previously investigated. The presented study aimed to explore the prospect of dapagliflozin to mitigate 2,4,6 trinitrobenzene sulfonic acid (TNBS)-induced rat colitis model which recapitulates several features of the human IBD. The molecular mechanisms pertaining to the dynamic balance between autophagy/apoptosis and colon injury were delineated, particularly, AMPK/mTOR, HMGB1/RAGE/NF-κB and Nrf2/HO-1 pathways. The colon tissues were examined using immunoblotting, ELISA, and histopathology. Dapagliflozin (0.1, 1 and 5 mg/kg; p.o.) dose-dependently mitigated colitis severity as manifested by suppression of the disease activity scores, macroscopic damage scores, colon weight/length ratio, histopathologic perturbations, and inflammatory markers. More important, dapagliflozin enhanced colonic autophagy via upregulating Beclin 1 and downregulating p62 SQSTM1 protein expression. In this context, dapagliflozin activated the AMPK/mTOR pathway by increasing the p-AMPK/AMPK and lowering the p-mTOR/mTOR ratios, thereby, favoring autophagy. Moreover, dapagliflozin dampened the colonic apoptosis via lowering the caspase-3 activity, cleaved caspase-3 expression, and Bax/Bcl-2 ratio. Furthermore, dapagliflozin attenuated the HMGB1/RAGE/NF-κB pathway via lowering HMGB1, RAGE, and p-NF-κBp65 protein expression. Regarding oxidative stress, dapagliflozin lowered the oxidative stress markers and augmented the Nrf2/HO-1 pathway. Together, the present study reveals, for the first time, the ameliorative effect of dapagliflozin against experimental colitis via augmenting colonic autophagy and curbing apoptosis through activation of AMPK/mTOR and Nrf2/HO-1 pathways and suppression of HMGB1/RAGE/NF-κB cascade.


Subject(s)
Apoptosis , Autophagy , Benzhydryl Compounds , Colitis, Ulcerative , Glucosides , Signal Transduction , Animals , Male , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Benzhydryl Compounds/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colon/pathology , Glucosides/therapeutic use , Heme Oxygenase (Decyclizing)/metabolism , HMGB1 Protein , NF-kappa B/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Trinitrobenzenesulfonic Acid , NF-E2-Related Factor 2
10.
Hum Exp Toxicol ; 40(5): 801-811, 2021 May.
Article in English | MEDLINE | ID: mdl-33118400

ABSTRACT

Public health issues have been raised regarding fructose toxicity and its serious metabolic disorders. Deleterious effects of high fructose intake on insulin sensitivity, body weight, lipid homeostasis have been identified. The new millennium has witnessed the emergence of a modern epidemic, the metabolic syndrome (MS), in approximately 25% of the world's adult population. The current study aimed to investigate the effect of the TNF-α antagonist infliximab on fructose-induced MS in rats. Rats were administered fructose (10%) in drinking water for 12 weeks to induce the experimental MS model. infliximab (5 mg/kg) was injected once weekly intraperitoneally starting on the 13th week for 4 weeks. Increase in body weight, blood glucose level, serum triglycerides (TGs), adiponectin level and blood pressure were present in MS rats. They also prompted increases in serum of leptin, TNF-α, and malondialdehyde (MDA) levels. Treatment with infliximab did not affect body weight, hyperglycemia or hypertension, but decreased serum TGs and increased serum HDL-c levels. Infliximab also decreased adiponectin levels. Surprisingly, infliximab increased MDA above its value in the MS group. These results reflect the fact that infliximab affects the manifestations of MS in rats. Though infliximab reduced TGs, increased HDL-c levels, reversed adiponectin resistance occurred by fructose, the drug failed to combat MS-mediated hyperglycemia, hypertension, and elevated MDA above the insult.


Subject(s)
Fructose/toxicity , Infliximab/metabolism , Infliximab/therapeutic use , Metabolic Syndrome/chemically induced , Metabolic Syndrome/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Middle Aged , Models, Animal , Rats , Rats, Sprague-Dawley
11.
Arch Biochem Biophys ; 693: 108552, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32860758

ABSTRACT

Ketoprofen is a widely used NSAID which incurs gastric mucosal damage. The high mobility group Box 1 (HMGB1) protein is a DNA-binding protein which exerts robust inflammatory actions, however, its role in ketoprofen-induced gastric damage has not been explored. Additionally, no previous studies have linked HMGB1/RAGE/NF-κB, DJ-1/Nrf2/HO-1 and PI3K/mTOR pathways in ketoprofen-induced gastropathy. The current work aimed to explore the potential of morin, a flavonoid with marked antioxidant/anti-inflammatory actions, to protect against ketoprofen-evoked gastric damage. Moreover, the underlying mechanisms, including the impact of morin on HMGB1/RAGE/NF-κB, DJ-1/Nrf2/HO-1 and PI3K/mTOR pathways were addressed. Immunoblotting and ELISA were used to examine the expression of target signals. Morin (50 mg/kg, p. o.) attenuated the severity of gastric injury via lowering of ulceration/hemorrhage and macroscopic damage scores. Meanwhile, it attenuated the histopathologic aberrations/damage scores. In the context of inflammation, morin suppressed TNF-α and myeloperoxidase levels and enhanced IL-10. Furthermore, it inhibited HMGB1/RAGE/NF-κB pathway through downregulating HMGB1, RAGE and phospho-NF-κBp65 protein expression. Morin successfully inhibited gastric mucosal oxidative stress through lowering of lipid peroxides and boosting of reduced glutathione, glutathione peroxidase and total antioxidant capacity. It also boosted DJ-1/Nrf2/HO-1 pathway via upregulating DJ-1, Nrf2 and HO-1 protein expression. Additionally, morin counteracted the apoptotic events by downregulating the proapoptotic Bax and Bax/Bcl-2 ratio and augmenting the PI3K/mTOR pathway through upregulating PI3Kp110α and phospho-mTOR protein expression. In conclusion, the current study demonstrates, for the first time, that morin shows a promise for the management of ketoprofen-induced mucosal insult through targeting of HMGB1/RAGE/NF-κB, DJ-1/Nrf2/HO-1 and PI3K/mTOR pathways.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Flavonoids/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Ketoprofen/pharmacology , Animals , Gastric Mucosa/injuries , HMGB1 Protein/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Male , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Deglycase DJ-1/metabolism , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Naunyn Schmiedebergs Arch Pharmacol ; 393(12): 2279-2292, 2020 12.
Article in English | MEDLINE | ID: mdl-32651660

ABSTRACT

Metabolic syndrome (MetS) is a low-grade inflammation state that results from an interplay between genetic and environmental factors. The incidence of MetS among individuals with insulin resistance, dyslipidemia, elevated blood pressure, and obesity, which constitute the syndrome, is 40% in the Middle East. The absence of an approved therapeutic agent for MetS is one reason to investigate tocilizumab (TCZ), which might be effective in the treatment of MetS. Results have implicated interleukin 6 (IL-6) in the development of MetS, identifying inflammation as a critical factor in its etiology and offering hope for new therapeutic approaches development. Here, we evaluate whether tocilizumab can be used for metabolic syndrome treatment. We assigned rats to three groups, 8 rats each: a negative-control group, provided with standard rodent chow and water; a fructose-fed group, provided with standard rodent chow and 10% fructose in drinking water for 22 weeks; and a treatment group, fed as per the metabolic syndrome group but treated with tocilizumab (5 mg/kg/week, intraperitoneal) for the final 5 weeks. Treatment with TCZ successfully ameliorated the damaging effects of fructose by stabilizing body weight gain and through the normalization of serum biochemical parameters and histopathological examination. Significant differences in adipokine levels were perceived, resulting in a significant decline in serum leptin and interleukin 6 (IL-6) levels concurrent with adiponectin normalization. Tocilizumab might be an effective agent for the treatment of metabolic syndrome. However, further investigations on human subjects are needed before the clinical application of tocilizumab for this indication.


Subject(s)
Adipokines/blood , Antibodies, Monoclonal, Humanized/therapeutic use , Fructose/toxicity , Interleukin-6/blood , Metabolic Syndrome/blood , STAT3 Transcription Factor/blood , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Insulin Resistance/physiology , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/prevention & control , Rats , Rats, Wistar
13.
Toxicol Appl Pharmacol ; 394: 114956, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32171571

ABSTRACT

Proper enterocytic proliferation/differentiation, besides providing adequate adherens junctions (AJ) integrity, are responsible for strengthening of the gut barrier that acts as a first line defense against endotoxemia. However, the preferential role of the underlying PI3K/Akt (PKB) axis in triggering enterocytic proliferation/differentiation signaling and AJ assembly is still obscure in sepsis. Additionally, the potential involvement of dipeptidyl peptidase (DPP)-IV in cholestatic sepsis has not yet been reported. Common bile duct ligation (CBDL) insult was performed in adult male Sprague-Dawley rats except for sham operated animals; three doses of vildagliptin (VLD3, 10 and 30 mg/kg/d; p.o) were administered for 10 consecutive days post CBDL. VLD3/10/30 dose-dependently decreased DPP-IV and elevated GLP-1, IGF-1, PI3K, pS473-Akt (PKB), pS9-GSK-3ß, pS133-CREB and cyclin-D1. VLD3/10 reduced fever, portal/aortic endotoxin and IgG, body weight loss as well as ileal NF-κB, TNF-α, MPO, TBARS, subepithelial/pericryptal and submucosal collagen deposition, vimentin immunoreactivity, N-cadherin, Zeb1 and pY654-ß-catenin but increased E-cadherin, NPSH and colon/spleen indices - effects that were quite the opposite of VLD30. Accordingly, maintaining proper enterocytic proliferation/differentiation and phosphorylation inputs consequent to adequate DPP-IV inhibition is integral to AJ assembly in cholestatic sepsis; however, perturbed signals by excessive suppression of the enzyme activity induce toxic effects manifested as AJ disassembly and EMT, hence gut leakage and overt endotoxemia.


Subject(s)
Cholestasis/pathology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Enterocytes/drug effects , Epithelial-Mesenchymal Transition/drug effects , Sepsis/pathology , Animals , Cell Differentiation/drug effects , Cholestasis/drug therapy , Common Bile Duct/injuries , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Gene Expression/drug effects , Ileum/pathology , Ligation , Male , Rats , Rats, Sprague-Dawley , Sepsis/drug therapy , Vildagliptin/pharmacology , Vimentin/metabolism , Weight Loss/drug effects
14.
Inflammation ; 43(2): 401-416, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31863220

ABSTRACT

Ischemic stroke is a major cause of death and motor disabilities all over the world. It is a muti-factorial disorder associated with inflammatory, apoptotic, and oxidative responses. Nateglinide (NAT), an insulinotropic agent used for the treatment of type 2 diabetes mellitus, recently showed potential anti-inflammatory and anti-apoptotic effects. The aim of our study was to elucidate the unique neuroprotective role of NAT in the middle cerebral artery occlusion (MCAO)-induced stroke in rats. Fifty-six male rats were divided to 4 groups (n = 14 in each group): the sham-operated group, sham receiving NAT (50 mg/kg/day, p.o) group, ischemia/reperfusion (IR) group, and IR receiving NAT group (50 mg/kg/day, p.o). MCAO caused potent deficits in motor and behavioral functions of the rats. Significant increase in inflammatory and apoptotic biomarkers has been observed in rats' hippocampi. Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway was significantly stimulated causing activation of series inflammatory biomarkers ending up neuro-inflammatory milieu. Pretreatment with NAT preserved rats' normal behavioral and motor functions. Moreover, NAT opposed the expression of hypoxia-inducible factor-1α (HIF-1α) resulting in downregulation of more inflammatory mediators namely, NF-κB, tumor necrosis factor-ß (TNF-ß), and the anti-survival gene PMAIP-1. NAT stimulated caveolin-1 (Cav-1) which prevented expression of oxidative biomarkers, nitric oxide (NO), and myeloperoxidase (MPO) and hamper the activation of apoptotic biomarker caspase-3. In conclusion, our work postulated that NAT exhibited its neuroprotective effects in rats with ischemic stroke via attenuation of different unique oxidative, apoptotic, and inflammatory pathways.


Subject(s)
Brain Ischemia/metabolism , Caveolin 1/biosynthesis , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nateglinide/therapeutic use , Receptors, Cell Surface/metabolism , Animals , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Down-Regulation/drug effects , Down-Regulation/physiology , Hippocampus/drug effects , Hippocampus/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Nateglinide/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rats , Rats, Wistar , Receptors, Cell Surface/antagonists & inhibitors , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Parasit Vectors ; 12(1): 304, 2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31208446

ABSTRACT

BACKGROUND: Schistosomiasis is responsible for a considerable global disease burden. This work aimed to improve the therapeutic outcome of the only available antischistosomal drug worldwide, praziquantel (PZQ), by incorporating it into a novel carrier, "solid lipid nanoparticles (SLNs)", to enhance its solubility, bioavailability and efficacy. A simple, cost-effective method was used to prepare SLN-PZQ. RESULTS: Compared to market PZQ (M-PZQ), SLN-PZQ was more bioavailable, as denoted by higher serum concentrations in both normal and infected mice where elevated Ka, AUC0-24, Cmax, and t1/2e with a decrease in kel were demonstrated. The AUC0-24 for SLN-PZQ in normal and Schistosoma mansoni-infected groups was almost nine- and eight-fold higher, respectively, than that for M-PZQ in corresponding groups. In normal and S. mansoni-infected mice, SLN-PZQ was detectable in serum at 24 h, while M-PZQ completely vanished 8 h post-treatment. Additionally, enhanced absorption with extended residence time was recorded for SLN-PZQ. Compared to M-PZQ, SLN-PZQ revealed superior antischistosomal activity coupled with enhanced bioavailability in all treated groups where higher percentages of worm reduction were recorded with all dosages tested. This effect was especially evident at the lower dose levels. The ED95 of SLN-PZQ was 5.29-fold lower than that of M-PZQ, with a significantly higher reduction in both the hepatic and intestinal tissue egg loads of all treated groups and almost complete disappearance of immature deposited eggs (clearly evident at the low dose levels). CONCLUSIONS: SLN-PZQ demonstrated enhanced PZQ bioavailability and antischistosomal efficacy with a safe profile despite the prolonged residence in the systemic circulation.


Subject(s)
Praziquantel/pharmacokinetics , Praziquantel/therapeutic use , Schistosomiasis mansoni/drug therapy , Schistosomicides/therapeutic use , Animals , Biological Availability , Drug Carriers/chemistry , Lipids/chemistry , Male , Mice , Nanoparticles/chemistry , Schistosoma mansoni/drug effects , Schistosomicides/pharmacokinetics
16.
J Mol Neurosci ; 68(1): 78-90, 2019 May.
Article in English | MEDLINE | ID: mdl-30863991

ABSTRACT

Long-term memory impairment is reported in more than 50% of cardiac arrest survivors. Monosialoganglioside (GM1) provided neuroprotection in experimental models of stroke but failed to replicate its promise clinically for unknown reasons. GM1 stimulates the release of nerve growth factor (NGF), which is synthesized as a precursor protein (pro-NGF) that either mediates apoptosis through the p75 neurotrophin receptor (p75NTR) or is cleaved by the protease furin (FUR) to yield mature NGF, the latter supporting survival through tropomyosin kinase receptor (Trk). The flavanol epicatechin (EPI) inhibits p75NTR-mediated signaling and apoptosis by pro-NGF. The aim of the current work is to test whether these two drugs affect, or communicate with, each other in the setting of CNS injuries. Using the two-vessel occlusion model of global ischemia/reperfusion (I/R), we tested if pharmacological modulation of Trk, p75NTR, and NGF balance with GM1, EPI, and their combination, can correct the memory deficit that follows this insult. Finally, we tested if FUR insufficiency and/or p75NTR-mediated apoptosis negatively affect the neurotherapeutic effect of GM1. Key proteins for Trk and p75NTR, FUR, and both forms of NGF were assessed. All treatment regiments successfully improved spatial memory retention and acquisition. A week after the insult, most Trk and p75NTR proteins were normal, but pro/mature NGF ratio remained sharply elevated and was associated with the poorest memory performance. Pharmacological correction of this balance was achieved by reinforcing Trk and p75NTR signaling. GM1 increased FUR levels, while concomitant administration of EPI weakened GM1 effect on pro-survival Trk and p75NTR mediators. GM1 neuroprotection is therefore not limited by FUR but could be dependent on p75NTR. Graphical Abstract "."


Subject(s)
Brain Ischemia/drug therapy , Memory Disorders/drug therapy , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Catechin/administration & dosage , Catechin/pharmacology , Catechin/therapeutic use , G(M1) Ganglioside/administration & dosage , G(M1) Ganglioside/pharmacology , G(M1) Ganglioside/therapeutic use , Male , Memory Disorders/etiology , Nerve Tissue Proteins , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Receptors, Growth Factor , Receptors, Nerve Growth Factor/metabolism
17.
Inflammation ; 42(3): 1056-1070, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30737662

ABSTRACT

The most epidemic liver disorder non-alcoholic steatohepatitis (NASH) is characterized by hepatic steatosis and inflammation with hepatocellular damage. Recently, it is predictable to be the extensive cause for liver transplantation. The absence of an approved therapeutic agent for NASH is the reason for investigating saroglitazar (SAR) which showed promising effects as a dual PPAR-α/γ agonist in recent studies on NASH. Here, we aimed to investigate the effect of SAR on NASH induced in rats by the administration of high-fat emulsion (HFE) and small doses of lipopolysaccharides (LPS) for 5 weeks. Rats were divided into three groups: negative control group (saline and standard rodent chow), model group (HFE(10 ml/kg/day, oral gavage) + LPS(0.5 mg/kg/week, i.p)), and SAR-treated group (HFE(10 ml/kg/day, oral gavage) + LPS(0.5 mg/kg/week, i.p.) + SAR(4 mg/kg/day, oral gavage) starting at week 3.Treatment with SAR successfully ameliorated the damaging effects of HFE with LPS, by counteracting body weight gain and biochemically by normalization of liver function parameters activity, glucose, insulin, homeostasis model of assessment (HOMA-IR) score, lipid profile levels, and histopathological examination. Significant changes in adipokine levels were perceived, resulting in a significant decline in serum leptin and tumor necrosis factor-α (TNF-α) level concurrent with adiponectin normalization. The positive effects observed for SAR on NASH are due to the downregulation of the LPS/TLR4 pathway, as indicated by the suppression of hepatic Toll-like receptor 4 (TLR4), NF-κB, TNF-α, and transforming growth factor-ß1 (TGF-ß1) expression. In conclusion, this work verified that SAR ameliorates NASH through deactivation of the hepatic LPS/TLR4 pathway and inhibition of adipocyte dysfunction.


Subject(s)
Adipocytes/drug effects , Liver/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Phenylpropionates/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Adipocytes/pathology , Animals , Down-Regulation , Lipopolysaccharides , Phenylpropionates/therapeutic use , Pyrroles/therapeutic use , Rats
18.
Life Sci ; 218: 284-291, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30611783

ABSTRACT

BACKGROUND AND PURPOSE: Rheumatoid arthritis (RA) is a chronic, systemic autoimmune inflammatory disease which poses a need to explore effective yet safe pharmacotherapeutic options. The current work aimed to study the therapeutic role of nicorandil in controlling RA. EXPERIMENTAL APPROACH: Complete Freund's adjuvant (CFA)-induced arthritis model was applied by injecting 400 µL of CFA in the right hind paw at day 0 and day 7. Four groups of rats were used as follows: normal-control (CTRL), CFA-induced arthritis (ART), CFA-induced arthritis treated with diclofenac (DIC) and CFA-induced arthritis treated with nicorandil (NIC). Both NIC and DIC were administered at day 14 for two weeks. Paw volume, knee joint diameter, pain behavior assessment as well as body weight were all periodically recorded throughout the experimental period. Following the sacrifice of animals at day 28, gene expressions of TLR-4, MyD88 and TRAF6 as well as extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), nuclear factor Kappa B (NF-κB) were quantified in hind paws tissue. Finally, the serum levels of the inflammatory biomarkers (tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) together with the histopathological examination of sections in the rat hind paw were recorded. RESULTS: Both NIC and DIC proved promising anti-arthritic potential mediated, at least in part through switching off TLR4-MyD88-TRAF6 axis as well as downstream TRAF6 dependent activated MAP kinases and NF-κB. CONCLUSION AND IMPLICATIONS: Nicorandil, via interfering with TLR4 signaling, sheds light on a potential clinical role of the drug in pursuit for safe and effective regimens for RA.


Subject(s)
Arthritis, Experimental/drug therapy , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Myeloid Differentiation Factor 88/metabolism , Nicorandil/pharmacology , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism , Adjuvants, Immunologic/toxicity , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Male , Myeloid Differentiation Factor 88/genetics , Pain Measurement , Rats , Rats, Wistar , Signal Transduction/drug effects , TNF Receptor-Associated Factor 6/genetics , Toll-Like Receptor 4/genetics , Vasodilator Agents/pharmacology
19.
Sci Rep ; 8(1): 16814, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30429582

ABSTRACT

Zymosan, a natural compound, provokes acute peritonitis and multiple organ dysfunction that affects the kidney, beside other organs via exaggerated inflammatory response. The aim of the present study is to test the role of cholinergic anti-inflammatory pathway (CAP) in alleviating acute kidney injury (AKI) induced by zymosan in BALB/c mice, using galantamine, a cholinesterase inhibitor, known to act via α7 nicotinic acetylcholine receptor (α7 nAChR) to stimulate CAP. Galantamine verified its anti-inflammatory effect by elevating acetylcholine (ACh) level, while abating the interleukin-6/ janus kinase 2 (Y1007/1008)/ signal transducer and activator of transcription 3 (Y705) (IL-6/ pY(1007/1008)-JAK2/ pY705-STAT3) inflammatory axis, with a consequent inhibition in suppressor of cytokine signaling 3 (SOCS3). This effect entails also the nuclear factor-kappa B (p65)/ high mobility group box protein-1/ (NF-κB (p65)/ HMGB-1) signaling pathway. Furthermore, the reno-curattive effect of galantamine was associated by a reduction in plasma creatinine (Cr), cystatin (Cys)-C, IL-18, and renal neutrophil gelatinase-associated lipocalin (NGAL), as well as an improved histopathological structure. Blocking the α7 nAChR by methyllycaconitine abolished the beneficial effect of galantamine to document the involvement of this receptor and the CAP in the amelioration of AKI induced by zymosan.


Subject(s)
Acute Kidney Injury/prevention & control , Zymosan/adverse effects , alpha7 Nicotinic Acetylcholine Receptor/physiology , Acetylcholine/metabolism , Aconitine/analogs & derivatives , Aconitine/pharmacology , Acute Kidney Injury/chemically induced , Animals , Cholinesterase Inhibitors/pharmacology , Galantamine/pharmacology , Inflammation/chemically induced , Mice , Mice, Inbred BALB C , Signal Transduction , alpha7 Nicotinic Acetylcholine Receptor/metabolism
20.
J Enzyme Inhib Med Chem ; 33(1): 1472-1493, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30191744

ABSTRACT

In this work, design, synthesis, and screening of thiophene carboxamides 4-13 and 16-23 as dual vascular endothelial growth factor receptors (VEGFRs) and mitotic inhibitors was reported. All compounds were screened against two gastrointestinal solid cancer cells, HepG-2 and HCT-116 cell lines. The most active cytotoxic derivatives 5 and 21 displayed 2.3- and 1.7-fold higher cytotoxicity than Sorafenib against HepG-2 cells. Cell cycle and apoptosis analyses for compounds 5 and 21 showed cells accumulation in the sub-G1 phase, and cell cycle arrest at G2/M phase. The apoptotic inducing activities of compounds 5 and 21were correlated to the elevation of p53, increase in Bax/Bcl-2 ratio, and increase in caspase-3/7.Compounds 5 and 21 showed potent inhibition againstVEGFR-2 (IC50 = 0.59 and 1.29 µM) and ß-tubulin polymerization (73% and 86% inhibition at their IC50 values).Molecular docking was performed with VEGFR-2 and tubulin binding sites to explain the displayed inhibitory activities.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Design , Drug Evaluation, Preclinical , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Thiophenes/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Quantitative Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...