Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Intervalo de año de publicación
1.
Iran J Basic Med Sci ; 27(3): 335-342, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38333751

RESUMEN

Objectives: Due to the crucial role of polyamines during fetal growth and development, we aimed to determine the effect of prenatal administration of agmatine, an endogenous active metabolite of arginine, and a nutritional supplement, on autistic-like behaviors, oxidative-anti-oxidative profile, and histopathological changes of the prefrontal cortex (PFC) and CA1 area of the hippocampus in valproic acid (VPA) model of autism in male rats. Materials and Methods: VPA was injected intraperitoneally on embryonic days (ED) 12.5, and the pregnant rats were gavaged with agmatine between E6.5 to E18.5 (13 days), at doses of 0.001, 0.01, and 0.1 mg/kg. The autism-like behaviors and memory of male pups were analyzed via open-field, three-chamber, and novel object recognition tests. Serum oxidative stress and the histological changes in the PFC and CA1 were assessed at the end of the study. Results: The results suggest that prenatal agmatine reduced autistic-like behaviors by decreasing cell loss in CA1 and PFC. We observed no alterations in superoxide dismutase (SOD) level and total anti-oxidant capacity (TAC) between groups. VPA decreased catalase (CAT) activities, while agmatine decreased malondialdehyde (MDA) activity. Conclusion: Overall, this investigation suggests that agmatine may be a potential candidate for the early treatment and even prevention of appearance of autism symptoms.

3.
Int J Neurosci ; : 1-13, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37815366

RESUMEN

OBJECTIVE: Despite significant advances that have been made in the treatment of traumatic brain injury (TBI), it remains a global health issue. This study aimed to investigate the synergistic effects of 17-ß estradiol (E2) and auraptene (AUR) on TBI treatment. METHODS: In total, 70 adult male Wistar rats were divided randomly into ten main groups: Sham, TBI, TBI + DMSO, TBI + AUR (4 mg/kg), TBI + AUR (8 mg/kg), TBI + AUR (25 mg/kg), TBI + E2 group, TBI + AUR (4 mg/kg) + E2 group, TBI + AUR (8 mg/kg) + E2 group and TBI + AUR (25 mg/kg) + E2 group. Diffuse TBI was caused by the Marmarou process in male rats. The brain's tissues were harvested to check the parameters of oxidative stress and levels of inflammatory cytokine. RESULTS: The finding revealed that TBI induced a significant increase in brain edema, pro-inflammatory cytokines and oxidant levels [MDA and NO], and also a decrease in the brain's antioxidant biomarkers [GPx, SOD]. We also found that E2 and AUR (25 mg/kg) significantly preserved the levels of these biomarkers. The combination of AUR concentrations and E2 showed that this treatment efficiently preserved the levels of these biomarkers. Furthermore, the combination of E2 and AUR (25 mg/kg) c could cause the most effective synergistic interaction. CONCLUSION: AUR could act synergistically with E2 to treat brain injury complications.

4.
Brain Behav ; 13(11): e3244, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37661235

RESUMEN

BACKGROUND: Studies have confirmed the salutary effects of progesterone (P4) on traumatic brain injury (TBI). This study investigated the beneficial effects of P4 via its receptors on TBI, and also whether progesterone receptors (PRs) can modulate TBI through PI3K/Akt pathway. MATERIAL AND METHODS: Marmarou method was utilized to induce diffuse TBI in ovariectomized rats. P4 (1.7 mg/kg) or the vehicle (oil) was administered 30 min after TBI induction. Moreover, RU486 (PR antagonist) and its vehicle (DMSO) were injected before TBI induction and P4 injection. Brain Evans blue content, brain water content (WC), various oxidative stress parameters, IL-1ß levels, tumor necrosis factor-α (TNF-α), histopathological alterations, and also phosphorylated Akt (p-Akt) and PI3K expressions in the brain were assessed 24 h after TBI. The veterinary comma scale (VCS) was measured before and after TBI at different times. RESULTS: The findings revealed that P4 caused an increase in VCS and a decrease in brain WC, oxidative stress, TNF-α and IL-1ß levels. RU486 inhibited the beneficial effects of P4 on these indices. Moreover, RU486 prevented the reduction of brain edema, inflammation, and apoptosis caused by P4. Moreover, P4 following TBI increased the expression of PI3K/p-Akt protein in the brain. RU486 eliminated the effects of P4 on PI3K/p-Akt expression. CONCLUSION: According to these findings, PRs are acting as critical mediators for the neuroprotective properties of P4 on oxidative stress, pro-inflammatory cytokine levels, and neurological outcomes. PRs also play an important role in regulating the PI3K/p-Akt expression and nongenomic function of P4.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fármacos Neuroprotectores , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Progesterona , Fosfatidilinositol 3-Quinasas/metabolismo , Fármacos Neuroprotectores/farmacología , Factor de Necrosis Tumoral alfa , Mifepristona/farmacología , Lesiones Traumáticas del Encéfalo/metabolismo , Progesterona/farmacología
5.
Prog Biophys Mol Biol ; 180-181: 19-27, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37080435

RESUMEN

Natural substances are increasingly being used as cancer treatments. Scutellarin, as a flavonoid, recently has been identified in a Chinese herbal extract called Erigeron breviscapus (Vant.). Scutellarin is being researched for its potential benefits due to the discovery that it possesses a variety of biological effects, such as neuroprotective, anti-bacterial, and anti-viral properties. In addition to these biological functions, scutellarin has also been found to have anti-tumor properties. The underlying mechanisms of scutellarin's anticancer activity involve its ability to inhibit various signaling pathways, such as Jak/STAT, ERK/AMPK, and Wnt/ß-catenin. Additionally, scutellarin activates intrinsic and extrinsic apoptotic pathways, which causes the death of tumor cells, interrupts the cell cycle, and promotes its arrest. By limiting metastasis, angiogenesis, drug resistance, and other tumorigenic processes, scutellarin also reduces the aggressiveness of tumors. Despite its promising anticancer activity, scutellarin faces several challenges in its clinical development, including poor solubility, bioavailability, and pharmacokinetic properties. Therefore, it has been suggested that certain modifications can enhance the pharmacogenetic capabilities of scutellarin to decrease its limited water solubility. In conclusion, scutellarin represents a potential candidate for cancer treatment and further studies are needed to explore its clinical utility and optimize its therapeutic potential.


Asunto(s)
Neoplasias , Extractos Vegetales , Transducción de Señal , Apigenina/farmacología , Apigenina/uso terapéutico , Medicina Tradicional , Neoplasias/tratamiento farmacológico
6.
Neurotoxicology ; 96: 154-165, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36933665

RESUMEN

Although anxiety disorders, as well as difficulties in social interaction, are documented in children with autism spectrum disorder (ASD) as a neurodevelopmental disorder, the effectiveness of potential therapeutic procedures considering age and sex differences is under serious discussion. The present study aimed to investigate the effect of resveratrol (RSV) on anxiety-like behaviors and social interaction in juvenile and adult rats of both sex in a valproic acid (VPA)-induced autistic-like model. Prenatal exposure to VPA was associated with increased anxiety, also causing a significant reduction in social interaction in juvenile male subjects. Further administration of RSV attenuated VPA-induced anxiety symptoms in both sexes of adult animals and significantly increased the sociability index in male and female juvenile rats. Taken together, it can be concluded that treatment with RSV can attenuate some of the harsh effects of VPA. This treatment was especially effective on anxiety-like traits in adult subjects of both sexes regarding their performance in open field and EPM. We encourage future research to consider the sex and age-specific mechanisms behind the RSV treatment in the prenatal VPA model of autism.


Asunto(s)
Trastorno del Espectro Autista , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Ratas , Femenino , Masculino , Animales , Ácido Valproico/efectos adversos , Resveratrol/farmacología , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ansiedad/inducido químicamente , Ansiedad/tratamiento farmacológico , Modelos Animales de Enfermedad , Conducta Social , Conducta Animal
7.
Sci Rep ; 13(1): 4780, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36959464

RESUMEN

Traumatic brain injury (TBI) causes progressive dysfunction that induces biochemical and metabolic changes that lead to cell death. Nevertheless, there is no definitive FDA-approved therapy for TBI treatment. Our previous immunohistochemical results indicated that the cost-effective natural Iranian medicine, Satureja khuzistanica Jamzad essential oil (SKEO), which consists of 94.16% carvacrol (CAR), has beneficial effects such as reducing neuronal death and inflammatory markers, as well as activating astrocytes and improving neurological outcomes. However, the molecular mechanisms of these neuroprotective effects have not yet been elucidated. This study investigated the possible mechanisms involved in the anti-inflammatory and anti-apoptotic properties of SKEO and CAR after TBI induction. Eighty-four male Wistar rats were randomly divided into six groups: Sham, TBI, TBI + Vehicle, TBI + CAR (100 and 200 mg/kg), and TBI + SKEO (200 mg/kg) groups. After establishing the "Marmarou" weight drop model, diffuse TBI was induced in the rat brain. Thirty minutes after TBI induction, SKEO & CAR were intraperitoneally injected. One day after TBI, injured rats exhibited significant brain edema, neurobehavioral dysfunctions, and neuronal apoptosis. Western blot results revealed upregulation of the levels of cleaved caspase-3, NFκB p65, and Bax/Bcl-2 ratio, which was attenuated by CAR and SKEO (200 mg/kg). Furthermore, the ELISA results showed that CAR treatment markedly prevents the overproduction of the brain pro-inflammatory cytokines, including IL-1ß, TNF-α, and IL-6. Moreover, the neuron-specific enolase (NSE) immunohistochemistry results revealed the protective effect of CAR and SKEO on post-TBI neuronal death. The current study revealed that the possible neuroprotective mechanisms of SKEO and CAR might be related to (at least in part) modulating NF-κB regulated inflammation and caspase-3 protein expression. It also suggested that CAR exerts more potent protective effects than SKEO against TBI. Nevertheless, the administration of SKEO and CAR may express a novel therapeutic approach to ameliorate TBI-related secondary phase neuropathological outcomes.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Encefalitis , Aceites Volátiles , Satureja , Ratas , Masculino , Animales , FN-kappa B/metabolismo , Aceites Volátiles/química , Satureja/química , Caspasa 3/metabolismo , Irán , Ratas Wistar , Lesiones Traumáticas del Encéfalo/patología , Inflamación/patología , Apoptosis , Encefalitis/metabolismo , Encéfalo/metabolismo
8.
Int J Neurosci ; : 1-14, 2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36379667

RESUMEN

OBJECTIVE: The benefits of exercise in TBI have been proven. However, the time-dependent effects of exercise initiation and the involved mechanisms are controversial. We investigated the effects of preconditioning, continuous, early, and delayed treadmill exercise on motor behavior, brain edema, inflammation, and oxidative stress in experimental traumatic brain injury (TBI). MATERIALS AND METHODS: 48 male rats were assigned into two groups: sedentary control (Sham and TBI) and exercise groups: 1MB (preconditioning, initiation beginning at 1 month before trauma), 1MBA (continuous, initiation beginning at 1 month before and continuing 1 month after trauma), 24hA (early, initiation beginning at 24 h after trauma), and 1WA (delay, initiation beginning at 1 week after trauma). The rats in exercise groups were forced to run on a treadmill five days a week for 30 min per day. Rotarod and open file were used to assess motor behavior. ELISA was also used to measure total antioxidant capacity (TAC), tumor necrosis factor-alpha (TNF-α), and malondialdehyde (MDA) in serum and CSF. RESULTS: Exercise significantly decreased neurological impairments, motor deficits, and apoptosis compared with the sedentary group. Early (within 24 h) and ongoing (1 MBA) exercise significantly improved motor behavior after TBI. In addition, these exercise programs inhibited brain edema and the number of apoptotic cells. MDA and TNF-α levels increased in all exercise groups, but the effects were greater after early exercise than after delayed exercise, resulting in a significant decrease in TAC levels in serum and CSF. We discovered a positive correlation between MDA, TAC, and TNF-α concentration in serum and CSF. CONCLUSION: Our finding suggests that early exercise (24hA) and 1MBA groups afford neuroprotection and reduce the second injury consequence, probably by reducing neuronal apoptosis and oxidative stress.

9.
Life Sci ; 305: 120744, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-35798069

RESUMEN

BACKGROUND: This study assessed the effects of single or combined administration of temozolomide (TMZ) and interferon-gamma (IFN-ᵞ) on anxiety-like behaviors, balance disorders, learning and memory, TNF-α, IL-10, some oxidant and antioxidants factors with investigating the toll-like receptor-4 (TLR4) and p-CREB signaling pathway in C6-induced glioblastoma of rats. METHODS: 40 male Sprague-Dawley rats bearing intra-caudate nucleus (CN) culture medium or C6 inoculation were randomly divided into five groups as follows: Sham, Tumor, TMZ, IFN-ᵞ and a TMZ + IFN-ᵞ combination. The open-field test (OFT), elevated plus maze (EPM), rotarod, and passive avoidance test (PAT) were done on days 14-17. On day 17 after tumor implantation, brain tissues were extracted for histopathological evaluation. TNF-α, IL-10, SOD, GPX, TAC, MDA, the protein level of TLR4 and p-CREB was measured. RESULTS: Combination therapy inhibited the growth of the tumor. Treatment groups alleviated tumor-induced anxiety-like behaviors and improved imbalance and memory impairment. SOD, GPX, and TAC decreased in the tumor group. The combination group augmented GPX and TAC. MDA decreased in treatment groups. TMZ, IFN-ᵞ reduced tumor-increased TNF-α and IL-10 level. The combination group declined TNF-α level in serum and IL-10 level in serum and brain. Glioblastoma induced significant upregulation of TLR4 and p-CREB in the brain which inhibited by IFN-ᵞ and TMZ+ IFN-ᵞ. CONCLUSION: The beneficial effects of TMZ, IFN-ᵞ, and TMZ+ IFN-ᵞ on neurocognitive functioning of rats with C6-induced glioblastoma may be mediated via modulating oxidative stress, reduced cytokines, and the downregulation of expression of TLR4 and p-CREB. Combination treatment appears to be more effective than single treatment.


Asunto(s)
Glioblastoma , Interferón gamma , Animales , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Interferón-alfa , Interferón gamma/farmacología , Interferón gamma/uso terapéutico , Interleucina-10 , Masculino , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa , Temozolomida/farmacología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
10.
Brain Res Bull ; 178: 108-119, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838851

RESUMEN

INTRODUCTION: The aim of this study was to determine the anti-inflammatory effect of female sex hormones on the level of intracellular molecules of cytokine signaling pathway after diffuse traumatic brain injury (TBI) in ovariectomized rats. METHODS: Female rats were divided into 10 groups: control, sham, TBI, Vehicle (oil), Vehicle E1 (33.3 µg/kg), E2 (1 mg / kg), P1 (1.7 mg/kg), P2 (8 mg / kg), E2 + P1. All drugs were injected 0.5 h after TBI. Brain edema and the brain levels of P-STAT-3, NFκB-P52, NFκB-P65, P-IκB, and SOCS-3 by immunohistochemistry measured at 24 h after TBI. RESULTS: Increased brain edema after TBI was inhibited by different doses of estrogen, progesterone (P < 0.001), and E2 + P1 (P < 0.05). The brain levels of P-STAT-3, NFκB-P52, NFκB-P65, and p-IκBα that increased after TBI was decreased only by E2 (P < 0.05). E2 and E2 + P1 have increased the SOCS-3 level after TBI (P < 0.05). Also, there was a difference between the E2 with E1 and two progesterone doses (P < 0.05). So that in all cases, the effects of E2 were more significant than the other groups. The target cells for these effects of E2 were microglia and astrocytes. CONCLUSION: The results indicate that one of the probable mechanism(s) of estrogen anti-inflammatory effect after TBI is either reduction of p-STAT-3, NFκB-P52, p-NFκB-P65, and p-IκBα or increase in SOCS-3 molecules involved in the signaling pathway of inflammatory cytokines.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Estrógenos/farmacología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Progesterona/farmacología , Animales , Modelos Animales de Enfermedad , Estrógenos/administración & dosificación , Femenino , Progesterona/administración & dosificación , Ratas
11.
Iran J Basic Med Sci ; 24(6): 760-766, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34630953

RESUMEN

OBJECTIVES: Along with increased intracranial pressure (ICP) and brain damage, brain edema is the most common cause of death in patients with hepatic encephalopathy. Curcumin can pass the blood-brain barrier and possesses anti-inflammatory and anti-oxidant properties. This study focuses on the curcumin protective effect on intrahepatic and extrahepatic damage in the brain. MATERIALS AND METHODS: One hundred and forty-four male Albino N-Mary rats were randomly divided into 2 main groups: intrahepatic injury group and extrahepatic cholestasis group. In intra-hepatic injury group intrahepatic damage was induced by intraperitoneal (IP) injection of acetaminophen (500 mg/kg) [19] and included four subgroups: 1. Sham, 2. Acetaminophen (APAP), 3. Normal saline (Veh) which was used as curcumin solvent, and 4. Curcumin (CMN). In extrahepatic cholestasis group intrahepatic damage was caused by common bile duct litigation (BDL) and included four subgroups: 1. Sham, 2. BDL, 3. Vehicle (Veh), and 4. Curcumin (CMN). In both groups, 72 hr after induction of cholestasis, brain water content, blood-brain barrier permeability, serum ammonia, and histopathological indicators were examined and ICP was measured every 24 hr for three days. RESULTS: The results showed that curcumin reduced brain edema, ICP, serum ammonia, and blood-brain barrier permeability after extrahepatic and intrahepatic damage. The maximum effect of curcumin on ICP was observed 72 hr after the injection. CONCLUSION: According to our findings, it seems that curcumin is an effective therapeutic intervention for treating encephalopathy caused by extrahepatic and intrahepatic damage.

12.
Brain Res Bull ; 177: 203-209, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34624461

RESUMEN

AIM: Traumatic Brain Injury (TBI) is widely acknowledged as a significant risk factor for death and disability. Our goal in this experiment was to see if Auraptene (AUR) could help rats recover from TBI-induced disability by measuring of oxidative stress parameters. MATERIAL AND METHODS: Adult male Wistar rats were randomly assigned to one of six groups: sham, TBI, Vehicle (DMSO), TBI+ AUR (4 mg/kg), TBI + AUR (8 mg/kg), TBI + AUR (25 mg/kg). The animals were anesthetized. After that, diffuse TBI was done by Marmarou model in male rats. Then, the brain tissues were harvested. Some of oxidative stress parameters, and TNFα levels were evaluated. RESULTS: TBI-induced brain damage was significantly inhibited by AUR (25 mg/kg), as evidenced by decreased Malondialdehyde (MDA) and Nitric Oxide (NO) levels, oxidative stress inhibition and reduced levels of pro-inflammatory cytokine tumor necrosis factor (TNF-α) in the brain. CONCLUSION: This study showed that probably the AUR prevents complications of TBI through decreases in brain edema, modulating oxidative stress, and reductions in the levels of inflammatory cytokines.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fármacos Neuroprotectores , Animales , Encéfalo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Cumarinas , Masculino , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo , Ratas , Ratas Wistar
13.
Life Sci ; 280: 119723, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34146552

RESUMEN

It has been shown that 17ß-estradiol (E2) hormone is an essential biological factor for increasing the sensitivity of women to drug abuse. Recent studies have shown a potential overlap between the molecular pathways of cannabinoids and ovarian hormones. The current study evaluated the interference between the marijuana and E2 effect on spatial learning and memory and the role of the G protein-coupled estrogen receptor (GPR30) in young female rats. The animals were separated into two main groups: intact-ovary and ovariectomized (OVX) rats. The latter group received intraperitoneal injections of E2, G-1 (GPR30 agonist), G-15 (GPR30 antagonist), marijuana, and different combinations of these substances for 28 days. Spatial learning and memory were evaluated by the Morris water maze (MWM) test. We also assessed the BDNF (brain-derived neurotrophic factor) concentration and the hippocampal level of GPR30. The results showed a significant reduction of spatial learning and memory in OVX rats compared to intact-ovary rats, which were restored by E2 replacement. Moreover, treatment with G-1 mimicked E2 effects on spatial learning and memory. Marijuana impaired spatial learning and memory in intact-ovary rats, while improved in OVX rats. We also found that treatment with M + E2 induced significant impairment in spatial learning and memory; however, treatment with M + G1 and M + G15 + E2 showed no significant difference. No significant differences in BDNF expression were observed in experimental groups. These results suggest that marijuana and E2 interact in their effect on spatial learning and memory in young female rats, but GPR30 seems to play no role in this interaction.


Asunto(s)
Cannabis/metabolismo , Estradiol/metabolismo , Extractos Vegetales/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Aprendizaje Espacial/efectos de los fármacos , Animales , Femenino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Extractos Vegetales/farmacología , Ratas , Ratas Wistar
14.
Iran J Basic Med Sci ; 24(3): 349-359, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33995946

RESUMEN

OBJECTIVES: The contribution of classic progesterone receptors (PR) in interceding the neuroprotective efficacy of progesterone (P4) on the prevention of brain edema and long-time behavioral disturbances was assessed in traumatic brain injury (TBI). MATERIALS AND METHODS: Female Wistar rats were ovariectomized and apportioned into 6 groups: sham, TBI, oil, P4, vehicle, and RU486. P4 or oil was injected following TBI. The antagonist of PR (RU486) or DMSO was administered before TBI. The brain edema and destruction of the blood-brain barrier (BBB) were determined. Intracranial pressure (ICP), cerebral perfusion pressure (CPP), and beam walk (BW) task were evaluated previously and at various times post-trauma. Long-time locomotor and cognitive consequences were measured one day before and on days 3, 7, 14, and 21 after the trauma. RESULTS: RU486 eliminated the inhibitory effects of P4 on brain edema and BBB leakage (P<0.05, P<0.001, respectively). RU486 inhibited the decremental effect of P4 on ICP as well as the increasing effect of P4 on CPP (P<0.001) after TBI. Also, RU486 inhibited the effect of P4 on the increase in traversal time and reduction in vestibulomotor score in the BW task (P<0.001). TBI induced motor, cognitive, and anxiety-like disorders, which lasted for 3 weeks after TBI; but, P4 prevented these cognitive and behavioral abnormalities (P<0.05), and RU486 opposed this P4 effect (P<0.001). CONCLUSION: The classic progesterone receptors have neuroprotective effects and prevent long-time behavioral and memory deficiency after brain trauma.

15.
Ecotoxicol Environ Saf ; 213: 111987, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33582408

RESUMEN

Protective effects of estrogen (E2) on traumatic brain injury (TBI) have been determined. In this study, the hepatoprotective effects of E2 after TBI through its receptors and oxidative stress regulation have been evaluated. Diffuse TBI induced by the Marmarou method in male rats. G15, PHTPP, MPP, and ICI182-780 as selective antagonists of E2 were injected before TBI. The results indicated that TBI induces a significant increase in liver enzymes [Alkaline phosphatase (ALP), Aspartate aminotransferase (AST), Alanine aminotransferase (ALT), Glutamyl transferase (GGT)], and oxidants levels [Malondialdehyde (MDA), Nitric oxide (NO)] and decreases in antioxidant biomarkers [Glutathione peroxidase (GPx) and Superoxide dismutase (SOD)] in the brain and liver, and plasma. We also found that E2 significantly preserved levels of these biomarkers and enzymatic activity. All antagonists inhibited the effects of E2 on increasing SOD and GPx. Also, the effects of E2 on brain MDA levels were inhibited by all antagonists, but in the liver, only ICI + G15 + E2 + TBI group was affected. The impacts of E2 on brain and liver and plasma NO levels were inhibited by all antagonists. The current findings demonstrated that E2 probably improved liver injury after TBI by modulating oxidative stress. Also, both classic (ERß, ERα) and non-classic [G protein-coupled estrogen receptor (GPER)] receptors are affected in the protective effects of E2.


Asunto(s)
Estradiol/farmacología , Sustancias Protectoras/farmacología , Alanina Transaminasa/metabolismo , Animales , Antioxidantes/metabolismo , Aspartato Aminotransferasas/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Glutatión Peroxidasa/metabolismo , Hígado/efectos de los fármacos , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Receptores de Estrógenos/metabolismo , Superóxido Dismutasa/metabolismo
16.
Biochem Pharmacol ; 178: 114044, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32445868

RESUMEN

Several studies have shown that 17ß-estradiol (E2) exerted beneficial effects on liver disease, and it has a protective impact on brain damage after traumatic brain injury (TBI). TBI-induced liver injury is associated with the activation of TLR4. However, it remains unknown whether E2 can modulate TBI-induced liver injury through TLR4. The objective of this study was to determine the role of TLR4 in hepatoprotective mechanisms of E2 after TBI. Diffuse TBI induced by the Marmarou model in male rats. TAK-242 as a selective antagonist of TLR4 (3 mg/kg) and E2 (33.3 µg/kg) were injected (i.p) respectively 30 min before and 30 min after TBI. The results showed that E2 and TAK-242 markedly inhibited TBI-induced liver injury, which was characterized by decreased aminotransferase activities, inhibition of the oxidative stress, and reduced levels of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and IL-17 in the liver. We also found that TBI induced significant upregulation of TLR4 in the liver, with peak expression occurring 24 h after TBI, and that treatment with E2 significantly inhibited the upregulation of TLR4. Also, both classic [Estrogen receptors alpha (ERα) and beta (ERß)] and non-classic (G protein-coupled estrogen receptor GPER) E2 receptors are involved in modulating the expression of TLR4. These results suggested that the hepatoprotective effects of estradiol after TBI may be mediated via the downregulation expression of TLR4.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Estradiol/uso terapéutico , Hígado/metabolismo , Receptor Toll-Like 4/fisiología , Animales , Lesiones Traumáticas del Encéfalo/patología , Estradiol/farmacología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Resultado del Tratamiento
17.
Brain Res Bull ; 140: 169-175, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29730418

RESUMEN

Brain edema and increased intracranial pressure (ICP) are among the main causes of neurological disturbance and mortality following traumatic brain injury (TBI). Since pregabalin neuroprotective effects have been shown, this study was performed to evaluate the possible neuroprotective effects of pregabalin in experimental TBI of male rats. Adult male Wistar rats were divided into 4 groups: sham, vehicle, pregabalin 30 mg/kg and pregabalin 60 mg/kg. TBI was induced in vehicle and pregabalin groups by Marmarou method. Pregabalin was administered 30 min after TBI. Sham and vehicle groups received saline. Brain water and Evans blue content and histopathological changes were evaluated 24, 5 and 24 h after TBI, respectively. The ICP and neurological outcomes (veterinary coma scale, VCS) were recorded before, 1 h and 24 h post TBI. The results showed a significant reduction in brain water content and ICP, and a significant increase in VCS of pregabalin group (60 mg/kg) as compared to vehicle group (P < 0.05). Also, pregabalin reduced brain edema and apoptosis score as compared to vehicle group. Post TBI pregabalin administration revealed a delayed but significant improvement in ICP and neurological outcomes in experimental TBI. The underlying mechanism(s) was not determined and needs further investigation.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Pregabalina/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Presión Intracraneal/efectos de los fármacos , Masculino , Ratas Wistar
18.
J. physiol. biochem ; 72(1): 33-44, mar. 2016. graf
Artículo en Inglés | IBECS | ID: ibc-168205

RESUMEN

The role of aquaporin-4 (AQP4) and interleukin-6 (IL-6) in the development of brain edema post-traumatic brain injury (TBI) has been indicated. The present study was designed to investigate the effect(s) of administration of progesterone (P) and/or estrogen (E) on brain water content, AQP4 expression, and IL-6 levels post-TBI. The ovariectomized rats were divided into 11 groups: sham, one vehicle, two vehicles, E1, E2, P1, P2, E1 + P1, E1 + P2, E2 + P1, and E2 + P2. The brain AQP4 expression, IL-6 levels, and water content were evaluated 24 h after TBI induced by Marmarou’s method. The low (E1 and P1) and high (E2 and P2) doses of estrogen and progesterone were administered 30 min post-TBI. The results showed that brain water content and AQP4 expression decreased in the E1, E2, P1, and P2-treated groups. The administration of E1 decreased IL-6 levels. Addition of progesterone decreased the inhibitory effect of E1 and E2 on the accumulation of water in the brain. Administration of E1 + P1 and E1 + P2 decreased the inhibitory effect of E1 on the IL-6 levels and AQP4 protein expression. Our findings suggest that estrogen or progesterone by itself has more effective roles in decrease of brain edema than combination of both. Possible mechanism may be mediated by the alteration of AQP4 and IL-6 expression. However, further studies are required to verify the exact mechanism (AU)


No disponible


Asunto(s)
Humanos , Estrógenos/administración & dosificación , Lesiones Encefálicas/metabolismo , Edema Encefálico/etiología , Progesterona/administración & dosificación , Acuaporina 4/metabolismo , Interleucina-6/metabolismo , Lesiones Encefálicas/complicaciones
19.
J Physiol Biochem ; 72(1): 33-44, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26638215

RESUMEN

The role of aquaporin-4 (AQP4) and interleukin-6 (IL-6) in the development of brain edema post-traumatic brain injury (TBI) has been indicated. The present study was designed to investigate the effect(s) of administration of progesterone (P) and/or estrogen (E) on brain water content, AQP4 expression, and IL-6 levels post-TBI. The ovariectomized rats were divided into 11 groups: sham, one vehicle, two vehicles, E1, E2, P1, P2, E1 + P1, E1 + P2, E2 + P1, and E2 + P2. The brain AQP4 expression, IL-6 levels, and water content were evaluated 24 h after TBI induced by Marmarou's method. The low (E1 and P1) and high (E2 and P2) doses of estrogen and progesterone were administered 30 min post-TBI. The results showed that brain water content and AQP4 expression decreased in the E1, E2, P1, and P2-treated groups. The administration of E1 decreased IL-6 levels. Addition of progesterone decreased the inhibitory effect of E1 and E2 on the accumulation of water in the brain. Administration of E1 + P1 and E1 + P2 decreased the inhibitory effect of E1 on the IL-6 levels and AQP4 protein expression. Our findings suggest that estrogen or progesterone by itself has more effective roles in decrease of brain edema than combination of both. Possible mechanism may be mediated by the alteration of AQP4 and IL-6 expression. However, further studies are required to verify the exact mechanism.


Asunto(s)
Acuaporina 4/metabolismo , Edema Encefálico/etiología , Lesiones Encefálicas/metabolismo , Estrógenos/administración & dosificación , Interleucina-6/metabolismo , Progesterona/administración & dosificación , Lesiones Encefálicas/complicaciones , Humanos
20.
Pharmacol Rep ; 66(1): 10-4, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24905300

RESUMEN

BACKGROUND: The role of transient receptor potential vanilloid 1 (TRPV1) in peripheral nervous system has been studied well but its role in the central nervous system remains to be studied in detail. The expression of TRPV1 receptors in hippocampus is suggesting that they may play an important role in higher cognitive functions such as learning and memory. METHODS: In the present study, the role of TRPV1 receptors in acquisition and retrieval of spatial memory of male Wistar rats was evaluated by intra-ventral hippocampus administration of TRPV1 selective agonist (OLDA) and antagonist (AMG9810) using Morris water maze. RESULTS: The results demonstrated that administration of either OLDA (0.001, 0.01 and 0.1 µg/rat) or AMG9810 (0.003, 0.03 and 0.3 µg/rat) did not influence memory acquisition or retrieval. CONCLUSIONS: These data suggest that ventral-hippocampal TRPV1 receptors possibly are not involved in spatial learning and memory.


Asunto(s)
Hipocampo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Canales Catiónicos TRPV/fisiología , Acrilamidas/farmacología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Dopamina/análogos & derivados , Dopamina/farmacología , Hipocampo/fisiología , Masculino , Ratas , Ratas Wistar , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...