Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Language
Publication year range
1.
Immunopharmacol Immunotoxicol ; 44(2): 168-177, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35021949

ABSTRACT

OBJECTIVE: Endoplasmic reticulum stress (ERS) and neuroinflammation are triggers for neurodegenerative disorders. Salubrinal is a selective inhibitor of protein phosphatase 1 (PP1) complex involving dephosphorylation of phosphorylated eukaryotic initiation factor-2α (eIF2α), the key crucial pathway in the ERS. Therefore, this study assessed the effects of inhibition of the ERS with salubrinal in the intranigral hemi-Parkinson disease (PD) model. MATERIALS AND METHODS: Animals were treated with salubrinal for one week after the PD model was created by intranigral lipopolysaccharide (LPS) administration. Apomorphine-induced rotation, rotarod, cylinder, and pole tests were performed to evaluate behavioral changes. Proinflammatory cytokines and the expression level of the dual specificity protein phosphatase 2 (DUSP2), PP1, and p-eIF2α were evaluated. Nigral expression of inducible nitric oxide synthase (iNOS), nuclear factor kappaB (Nf-κB), and cyclooxygenase (COX)-2 was determined. Finally, tyrosine hydroxylase and caspase-3/ caspase-9 expressions were assessed by immunohistochemistry. RESULTS: Salubrinal reduced the motor impairments and dopamine-related behavioral deficiencies caused by the LPS. Salubrinal attenuated the LPS-induced increased levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, and salubrinal rescued the loss of TH expression and dopamine levels and prevented the caspase-3/9 increase in the substantial nigra (SN). LPS potently increased iNOS, Nf-κB, and COX-2 expression, but this effect was reduced after salubrinal treatment. Additionally, salubrinal attenuated the LPS-induced PP1 and DUSP2 increase. CONCLUSION: Our results reveal that salubrinal is attenuating several inflammatory mediators and thereby decreased the inflammatory effects of LPS in the neurons of the SN. Together this results in increased cellular survival and maintained integrity of SN. Taken together our data show the beneficial effects of inhibition of ERS to restrict neuroinflammatory progression and neuronal loss in a PD model.


Subject(s)
Lipopolysaccharides , Parkinson Disease , Animals , Cinnamates , Lipopolysaccharides/toxicity , Microglia/metabolism , NF-kappa B/metabolism , Neuroinflammatory Diseases , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Substantia Nigra/metabolism , Thiourea/analogs & derivatives
2.
Braz. J. Pharm. Sci. (Online) ; 58: e20942, 2022. graf
Article in English | LILACS | ID: biblio-1420449

ABSTRACT

Abstract In the last decades, ferroptosis and its relationship with Parkinson's disease have gained significant attention. Compounds that affect ferroptosis and iron-dependent pathways in particular, have possible candidates for study in this context.Sinapic acid is an iron-chelator and high antioxidant bioactive phenolic acid. Its neuroprotective action, due to the antioxidant capacity, has been shown in several experimental models.However, the relationship between iron and antioxidant actions is still misunderstood and therefore, in the current study, we tried to investigate the effects of sinapic acid in rotenone-induced Parkinson's disease with the aspect of ferroptosis and iron-dependent alterations.The Parkinson's disease model was induced by a single dose intrastriatal and intrategmental rotenone (5µg/µl) injection.Sinapic acid (30mg/ kg) was orally administered during a 28-day period after the Parkinson's disease model was validated.Our results demonstrated that sinapic acid treatment attenuated rotenone-induced increase of serum transferrin and iron levels.Furthermore, sinapic acid inhibited rotenone-induced heme oxygenase-1(HO-1) increase and decrease of glutathione peroxidase-4 (GPx-4) levels in brain tissue. Also, sinapic acid treatment decreased motor impairment, likely as a result of the ameliorative effects on the tyrosine hydroxylase immunoreactivity loss after the rotenone insult.Our study suggests that the iron regulatory role of sinapic acid possibly plays a role in the protective effect on rotenone-induced neuronal damage.


Subject(s)
Animals , Male , Rats , Rotenone/adverse effects , Neuroprotective Agents/agonists , Iron/adverse effects , Ferroptosis
3.
Neurosci Lett ; 738: 135308, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32932183

ABSTRACT

Lenalidomide is a centrally active thalidomide analog that has potent anti-inflammatory and antiangiogenic activities. Currently, it is primarily used in the treatment of multiple myeloma and myelodysplastic syndromes. However, recent studies have revealed in addition to neuroprotection and neuromodulation of lenalidomide. Because of this combination of inflammation and neuro-immunogenic properties, lenalidomide is considered as a high potential compound for the treatment of neurodegenerative diseases. Despite intensive research during the last decade, the role of neurotrophic elements in the effect of lenalidomide is still not well understood. Therefore, in the current study, the effects of lenalidomide on neurodegeneration were investigated in a rotenone model of Parkinson's disease (PD) rat model. The PD rat model was generated by rotenone injection into the substantia nigra pars compacta (SNpc). After validation of the PD model, the rats were treated with lenalidomide (100 mg/kg) for 28 days. Our data shows that lenalidomide alleviated rotenone-induced motor impairments and deficits in dopamine-related behaviors and resulted in increased levels of tumor necrosis factor-α and calcium-binding protein B in the SNpc. Moreover, chronic lenalidomide treatment resulted increase in transforming growth factor immunoreactivity and brain derived neurotrophic factor expression in the SNPc. In addition, chronic treatment mitigated tyrosine hydroxylase expression prevented the rotenone-induced decrease in dopamine levels, and consequently a decrease in caspase-3/9 immunoreactivity. This thus shows that chronic lenalidomide treatment improves neuronal survival. Together with our data demonstrate that lenalidomide, in addition to its anti-inflammatory and immunomodulatory actions, is also capable of increasing neurotrophic factors in the SNpc, thereby preventing rotenone-induced motor impairments.


Subject(s)
Lenalidomide/therapeutic use , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Pars Compacta/drug effects , Rotenone , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Binding Proteins/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Lenalidomide/administration & dosage , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Pars Compacta/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/metabolism
4.
Balkan Med J ; 30(4): 355-61, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25207141

ABSTRACT

BACKGROUND: Alpha2 agonists contribute to pain control at the level of the medulla spinalis. Alpha2 agonists are generally added to local anaesthetics to prolong spinal or epidural anaesthesia time. AIMS: In the present study, we aimed to evaluate the antinociceptive and neurotoxic effects of dexmedetomidine given intracerebroventricularly for 5 days. STUDY DESIGN: Animal experimentation. METHODS: After intraventricular cannulation, rats (n=32) were divided into two groups (n=16 each). Rats in the dexmedetomidine group (Group D, n=16) received 3 µg (0.03 mL) dexmedetomidine and the control group (Group C, n=16) received 0.03 mL physiological serum through an intracerebroventricular catheter once a day, for 5 days. Antinociceptive, sedative, and motor effects were evaluated before the injection and for 90 min after injection. The tail-flick and hot plate tests were used to assess thermal nociceptive threshold. For histopathological evaluation, half of the rats in both groups were sacrificed on the 6(th) day and the remaining rats were sacrificed on the 21(st) day. Then the perfusion fixation method was applied. The first tissue section was obtained from the cervical spinal cord 1 cm distal to the proximal end of the spinal cord. The second sample was retrieved from the region 1 cm distal from the thoracic 13-lumbar 1 vertebra. On morphological evaluation, nonspecific changes like edema and gliosis, signs of neuronal degeneration demonstrating a severe reaction, and density of inflammatory cells were examined. RESULTS: In dexmedetomidine-administered rats, on the first day reaction times at 5, 10, and 20 min and on the other days, reaction times at 5, 10, 20, and 30 min in hot plate tests were significantly longer compared with baseline values (p<0.05). In dexmedetomidine-administered rats, on the 1(st), 4(th), and 5(th) days reaction times at 5, 10, 20, 30, and 40 min and on the 2(nd) and 3(rd) days reaction times at 5, 10, 20, and 30 min in tail-flick tests were significantly longer compared with baseline values (p<0.05). First-degree sedation lasting for 60 min and first-degree motor block lasting for 30-40 min were observed in the dexmedetomidine group. Similar rates of nonspecific changes such as edema and gliosis were seen in both groups. Signs of severe reactions such as neuronal degeneration and diffuse inflammatory cell infiltration were not encountered in any group. There was no significant difference between groups according to morphological findings of the spinal cord on the 6(th) and 21(st) days (p>0.05). CONCLUSION: We observed that intracerebroventricular administration of 3 µg dexmedetomidine produced antinociception and did not cause neurotoxicity.

SELECTION OF CITATIONS
SEARCH DETAIL
...