Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Behav Pharmacol ; 34(5): 263-274, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37401406

RESUMO

Temozolomide is used commonly in the treatment of some types of cancers, but it may also result in cognitive impairments such as memory deficits. l -Dopa, a well known medicine for the central nervous system, has been shown to have positive effects on some cognitive disorders. Here we sought to investigate the effect of l -Dopa on temozolomide-induced cognitive impairments. BALB/c mice were subjected to 3-days temozolomide and 6-days concomitant l -Dopa/benserazide administration in six groups (control, l -Dopa 25 mg/kg, l -Dopa 75 mg/kg, temozolomide, temozolomide +  l -Dopa 25 mg/kg, and temozolomide +  l -Dopa 75 mg/kg). Open field test, object location recognition, novel object recognition test, and shuttle-box test were carried out to determine the locomotor, anxiety-like behavior, and memory function of subjects. TNF-α and brain-derived neurotrophic factor (BDNF) gene expression in the hippocampus was measured by real-time PCR. Mice treated with temozolomide showed recognition memory impairment, along with hippocampal TNF-α and BDNF mRNA expression level raise, and detection of histological insults in hematoxylin and eosin hippocampal slides. Mice that received temozolomide +  l -Dopa showed normal behavioral function and lower TNF-α and BDNF hippocampal mRNA expression levels, and histologically normal hippocampal CA1 region in comparison with mice in the temozolomide group. Our results provide evidence that l -Dopa prevents temozolomide-induced recognition memory deficit in mice at the acute phase probably via l -Dopa antineuroinflammatory effects.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Disfunção Cognitiva , Camundongos , Masculino , Animais , Temozolomida/farmacologia , Temozolomida/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Hipocampo , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , RNA Mensageiro/metabolismo
2.
Exp Brain Res ; 240(9): 2339-2348, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35859208

RESUMO

Cerebellar ataxia is a neurodegenerative disorder leading to severe motor incoordination. Recently, it has been suggested that cannabinoids play a role in modulating ataxic symptoms. To understand the possible therapeutic effect of cannabinoids for the management of cerebellar ataxia, we used cannabinoid agonist/antagonists to target the cannabinoid type 1 receptor (CB1R) in the 3 acetyl pyridine (3AP) rat model of ataxia. The role of the CB1R was examined using three different doses of the CB1R agonist, WIN-55,212-2 (WIN; 0.1, 0.5, 1 mg/kg) administrated 30 min prior to 3AP (55 mg/kg, i.p.) which leads to motor impairment through destruction of the inferior olive. In some groups, the CB1R antagonist AM251 (1 mg/kg) was given in combination with WIN. Locomotor activity and motor coordination were impaired by 3AP, and the application of WIN did not ameliorate this effect. However, the abnormal gait, rearing and grooming caused by 3AP were prevented by co-administration of AM251 with WIN. While the addition of the CB1R antagonist improved some ataxic symptoms, there was no effect of AM251 on balance or locomotor activity when co-administrated with WIN. Behavioral testing indicated that not only did WIN fail to exert any protective effect on ataxic symptoms; it exacerbated ataxic symptoms, suggesting that CB1R agonists may not be the ideal therapeutic drug in this disorder. When taken together, the findings from the present study indicate that cannabinoid modulation of ataxia symptoms may not act solely through CB1Rs and other cannabinoid receptors should be considered in future studies.


Assuntos
Canabinoides , Ataxia Cerebelar , Animais , Agonistas de Receptores de Canabinoides/farmacologia , Agonistas de Receptores de Canabinoides/uso terapêutico , Canabinoides/farmacologia , Canabinoides/uso terapêutico , Ataxia Cerebelar/tratamento farmacológico , Piridinas/farmacologia , Piridinas/uso terapêutico , Ratos , Receptor CB1 de Canabinoide
3.
Int J Neurosci ; : 1-8, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36120979

RESUMO

Aim: Disruption in cerebellar inputs, as well as dysfunction of Purkinje cells (PCs), causes a change in the timing of electrical signaling in the cerebellum resulting in disorders such as cerebellar ataxia. Although much clinical and molecular genetics research has been conducted to understand this disorder, there is no specific treatment for cerebellar ataxia. As cannabinoid type 1 receptors (CB1Rs) are highly expressed in the cerebellum and have been suggested as a therapeutic strategy, we determined whether AM251, a cannabinoid receptor antagonist, was neuroprotective of PCs in a rat cerebellar ataxic model.Materials and methods: To this end, we conducted behavioral and histological tests in the 3-acetylpyridine (3AP) rat cerebellar ataxia model, to explore whether AM251 was protective against induction of ataxia and cell death.Results: Rats with chemical degeneration of the inferior olive induced by 3AP (55 mg/kg, i.p.) clearly showed cerebellar ataxic symptoms. The locomotor activity and motor coordination of the ataxic animals were clearly disrupted compared to the control group. Further, histological analysis showed cell death and PCs degenerated with loss of cell membrane integrity associated with 3AP. Pre-treatment by AM251 improved the locomotor activity of the ataxic animals, and AM251 almost prevented PCs neuronal degeneration.Conclusion: Our data which show protection of cerebellar PCs and motor improvement in the ataxic rat model by treatment with AM251 suggests that targeting cannabinoid receptors should be considered for therapeutic intervention in cerebellar ataxia. HIGHLIGHTS:AM251 was protective against induction of ataxia and cell death.CBR antagonist typically ameliorated 3AP induced Ataxia.AM251 affected explorative and gait disturbances induced by 3AP.CBR antagonist improved impairments of anxiety-like behaviors following 3AP.

4.
Int J Neurosci ; 130(9): 865-874, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31860371

RESUMO

Aim: Empathy is defined as the capability to comprehend and simulate the feelings of others. Though it has been considered as a human feature, recent studies have demonstrated empathy-like behaviors in other animals including rats. The objective of the current study was to evaluate the role of nitric oxide system in cognition and nociception changes following observation of cagemates in pain.Material and methods: Adult male Wistar rats were used (n = 8 for each group). One sibling received formalin injection into the hindpaw five times within a nine-day period and the other sibling observed the pain while being pretreated with saline, L-NAME or L-arginine (10 mg/kg, i.p.). Nociception, anxiety-like behavior and locomotion, balance, muscle strength, spatial and fear learning were evaluated.Results: Observing a family member (sibling) in pain increased anxiety-like behavior, led to a hyperalgesia in the observer and disruption of spatial memory. Nitric oxide system modulated these changes, so that in some paradigms the activation of NO and in some others inhibition of NO dampened the effect of observing pain in a cagemate on the evaluated features.Conclusions: Results in the current study demonstrated a modulating effect of NO on empathy induced changes in nociception, motor function and spatial memory. Further studies addressing the specific brain regions and other neurotransmitters involved are recommended.


Assuntos
Ansiedade , Comportamento Animal/fisiologia , Empatia/fisiologia , Hiperalgesia , Óxido Nítrico/metabolismo , Nociceptividade/fisiologia , Memória Espacial/fisiologia , Animais , Ansiedade/etiologia , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Arginina/farmacologia , Comportamento Animal/efeitos dos fármacos , Empatia/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nociceptividade/efeitos dos fármacos , Ratos , Ratos Wistar , Irmãos , Memória Espacial/efeitos dos fármacos
5.
Electromagn Biol Med ; 39(4): 298-309, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32666844

RESUMO

Thirteen million cancer deaths and 21.7 million new cancer cases are expected in the world by 2030. Glioblastoma is the most common primary malignant tumor of the central nervous system which is the most lethal type of primary brain tumor in adults with the survival time of 12-15 months after the initial diagnosis. Glioblastoma is the most common and most malignant type of brain tumor, and despite surgery, chemotherapy and radiation treatment, the average survival of patients is about 14 months. The current research showed that the frequency magnetic field (FMF) and static magnetic field (SMF) can influence cancer cell proliferation and coupled with anticancer drugs may provide a new strategy for cancer therapy. At the present study, we investigated the effects of FMF (10 Hz, 50 G), SMF (50 G) and Temozolomide (200 µm) on viability, free radical production, and p53 followed by p53 protein expression in the human glioblastoma cell line (A172) by MTT, NBT, RT-PCR and Western blot. Results showed that the effect of Temozolomide (TMZ) with SMF and FMF together increased the cytotoxicity, free radical production, and p53 followed by p53 protein expression in the human glioblastoma cell line (A172).


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Radicais Livres/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glioblastoma/patologia , Campos Magnéticos , Temozolomida/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Proteína Supressora de Tumor p53/genética
6.
Electromagn Biol Med ; 38(3): 198-209, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31179753

RESUMO

Glioblastoma multiforme (GBM) is a highly malignant brain tumor with an extremely dismal prognosis, a median survival is12 months. Temozolomide (TMZ) is an alkylating agent widely used to treat cancer, resistance to this drug is often found. One unexplored possibility for overcoming this resistance is a treatment based on concomitant exposure to electromagnetic fields (EMF) and TMZ. Indeed, many evidences show that EMF affects cancer cells and drug performance. Therefore, the present study was carried out to evaluate the potential synergistic effect of 100 µM TMZ and EMF (100 Hz, 100 G) on human glioma cell line U87 U87 cells with four experimental groups (I-IV) were exposed to ELF-EMF and TMZ for 120 and 144 h, as follows: (I) control; (II) ELF-EMF; (III) TMZ; (IV) ELF-PEMFs / TMZ. mRNA expression of genes such as (Nestin,CD133, Notch4 and GFAP) were investigated by Real-time PCR and western blot. We also evaluated, SOD activity, MDA and calcium concentration by ELISA assay. Co-treatment synergistically decreased the expression of Nestin,CD133, and Notch4 and increased the GFAP genes. We also observed an increase in Superoxide dismutase (SOD) activity, Malondialdehyde (MDA) and Ca2+concentration in comparison to controls.TMZ prevents cancer progression not only through the induction of cell death, but also by inducing differentiation in cancer cells. In addition, our data demonstrate ELF-EMF (100 Hz, 100 G) can significantly enhance the effects of TMZ on human glioblastoma U87 cell. These findings may open new window for future studies.


Assuntos
Antineoplásicos/farmacologia , Campos Eletromagnéticos , Glioblastoma/patologia , Temozolomida/farmacologia , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Cálcio/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/efeitos da radiação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Superóxido Dismutase/metabolismo , Temozolomida/administração & dosagem
7.
Int J Neurosci ; 128(8): 691-696, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29185809

RESUMO

INTRODUCTION: Although studies have shown a potential association between extremely low frequency electromagnetic fields (ELF-EMFs) exposure and Alzheimer's disease (AD), few studies have been conducted to investigate the effects of weak magnetic fields on brain functions such as cognitive functions in animal models. Therefore, this study aimed to investigate the effect of ELF-EMF exposure (50 Hz, 10 mT) on spatial learning and memory changes in AD rats. METHODS: Amyloid-ß (Aß) 1-42 was injected into lateral ventricle to establish an AD rat model. The rats were divided into six groups: Group I (control); Group II (surgical sham); Group III (AD) Alzheimer's rat model; Group IV (MF) rats exposed to ELF-MF for 14 consecutive days; Group V (Aß injection+M) rats exposed to magnetic field for 14 consecutive days from day 0 to 14 days after the Aß peptide injection; Group VI (AD+M) rats exposed to magnetic field for 14 consecutive days after 2 weeks of Aß peptide injection from 14th to 28th day . Morris water maze investigations were performed. RESULTS: AD rats showed a significant impairment in learning and memory compared to control rats. The results showed that ELF-MF improved the learning and memory impairments in Aß injection+M and AD+M groups. CONCLUSION: Our results showed that application of ELF-MF not only has improving effect on different cognitive disorder signs of AD animals, but also disrupts the processes of AD rat model formation.


Assuntos
Doença de Alzheimer/complicações , Magnetoterapia/métodos , Transtornos da Memória/etiologia , Transtornos da Memória/terapia , Recuperação de Função Fisiológica/efeitos da radiação , Peptídeos beta-Amiloides/toxicidade , Análise de Variância , Animais , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos da radiação , Fragmentos de Peptídeos/toxicidade , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/efeitos da radiação , Natação , Fatores de Tempo , Resultado do Tratamento
8.
Electromagn Biol Med ; 37(3): 138-145, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29846098

RESUMO

Glioblastoma multiforme (GBM) is a malignant brain cancer that causes high mortality in patients. GBM responds weakly to the common cancer treatments such as chemotherapy and radiotherapy and even surgery. Carboplatin is an alkylating agent widely used to treat cancer. However, resistance to this drug is a common problem in its use in cancer treatment. Concomitant exposure to extremely low-frequency electromagnetic fields (ELF-EMFs) and carboplatin is one unexplored possibility for overcoming this resistance. Indeed, many lines of evidence show that EMF affects cancer cells and drug action. In this study, we evaluated the effect of concomitant administration of carboplatin and EMF (50 Hz, 70 G) and also concomitant administration of carboplatin and static magnetic field (SMF) (70 G) on human glioma cell line (U-87). The results showed that cotreatment reduced the efficiency of carboplatin in U-87 cells, by decreasing caspase-3 in comparison to drug groups. Overall, EMF reduced the apoptotic effect of carboplatin, possibly through a redox regulation mechanism. Therefore, we have to avoid coadministration of magnetic field (MF) and carboplatin in tumor area, because the MF decreased the toxicity of the drug. However, further studies are needed to reveal the action mechanism of this combination therapeutic method.


Assuntos
Antineoplásicos/farmacologia , Carboplatina/farmacologia , Campos Eletromagnéticos , Glioblastoma/patologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos
9.
Curr Radiopharm ; 17(1): 55-67, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38817005

RESUMO

BACKGROUND: Exposure to physical contamination during chemotherapy, including non-ionizing electromagnetic fields, raises concerns about the widespread sources of exposure to this type of radiation. Glioblastoma multiforme (GBM) is an aggressive central nervous system tumor that is hard to treat due to resistance to drugs such as temozolomide (TMZ). OBJECTIVE: Electromagnetic fields (EMF) and haloperidol (HLP) may have anticancer effects. In this study, we investigated the effects of TMZ, HLP, and EMF on GBM cell lines and analyzed the association between non-ionizing radiation and the risk of change in drug performance. METHODS: Cell viability and reactive oxygen species (ROS) generation were measured by MTT and NBT assay, respectively. Then, the expression levels of breast cancer-resistant protein (BCRP), Bax, Bcl2, Nestin, vascular endothelial growth factor (VEGF) genes, and P53, Bax, and Bcl2 Proteins were evaluated by real-time PCR and western blot. RESULTS: Co-treatment of GBM cells by HLP and TMZ enhanced apoptosis in T-98G and A172 cells by increasing the expression of P53 and Bax and decreasing Bcl-2. Interestingly, exposure of GBM cells to EMF decreased apoptosis in the TMZ+HLP group. CONCLUSION: In conclusion, EMF reduced the synergistic effect of TMZ and HLP. This hypothesis that patients who are treated for brain tumors and suffer from depression should not be exposed to EMF is proposed in the present study. There appears to be an urgent need to reconsider exposure limits for low-frequency magnetic fields, based on experimental and epidemiological research, the relationship between exposure to non-ionizing radiation and adverse human health effects.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Apoptose , Sobrevivência Celular , Campos Eletromagnéticos , Haloperidol , Proteínas de Neoplasias , Nestina , Temozolomida , Fator A de Crescimento do Endotélio Vascular , Humanos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Nestina/metabolismo , Temozolomida/farmacologia , Haloperidol/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/biossíntese , Glioblastoma/radioterapia , Glioblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/metabolismo , Antineoplásicos Alquilantes/farmacologia , Glioma/radioterapia , Glioma/metabolismo , Glioma/patologia
10.
Curr Radiopharm ; 16(2): 123-132, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36503396

RESUMO

BACKGROUND: Glioblastoma is the most common primary malignant tumor of the central nervous system. The patient's median survival rate is 13.5 months, so it is necessary to explore new therapeutic approaches. OBJECTIVE: Extremely low-frequency electromagnetic field (EMF) has been explored as a noninvasive cancer treatment. This study applied the EMF with previous conventional chemoradiotherapy for glioblastoma. METHODS: In this study, we evaluated the cytotoxic effects of EMF (50 Hz, 100 G), temozolomide (TMZ), and radiation (Rad) on gene expression of T98 glioma cell lines in monolayer and spheroid cell cultures. RESULTS: Treatment with Rad and EMF significantly increased apoptosis-related gene expression compared to the control group in monolayers and spheroids (p<0.001). The expression of apoptotic-related genes in monolayers was higher than the similar spheroid groups (p<0.001). We found that treatment with TMZ and EMF could increase the gene expression of the autophagy cascade markers compared to the control group (p<0.001). Autophagy-related gene expression in spheroids was higher than in the similar monolayer group (p<0.001). We demonstrated that coadministration of EMF, TMZ, and Rad significantly reduced cell cycle and drug resistance gene expression in monolayers and spheroids (p<0.001) compared to the control group. CONCLUSION: The combinational use of TMZ, Rad and, EMF showed the highest antitumor activity by inducing apoptosis and autophagy signaling pathways and inhibiting cell cycle and drug resistance gene expression. Furthermore, EMF increased TMZ or radiation efficiency.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Campos Eletromagnéticos , Linhagem Celular Tumoral , Compostos Radiofarmacêuticos/farmacologia , Apoptose/efeitos da radiação , Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos
11.
Int J Pharm ; 637: 122884, 2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-36966981

RESUMO

According to the favorable antitumor properties of selenium, this study aimed to design a novel form of selenium nanoparticles (Se NPs) functionalized with chitosan (Cs) and sialic acid to assess their antitumor effects on the human glioblastoma cell lines (T98 and A172). Se NPs were synthesized in the presence of chitosan and ascorbic acid (Vc) and the synthesis conditions were optimized using response surface methodology. Se NPs@Cs were obtained with a monoclinic structure with an average diameter of 23 nm under the optimum conditions (reaction time = 30 min, chitosan concentration = 1 % w/v, Vc/Se molar ratio = 5). To modify Se NP@Cs for glioblastoma treatment, sialic acid was used to cover the surface of the NPs. Sialic acid was successfully attached to the surface of Se NPs@Cs, and Se NPs@Cs-sialic acid were formed in the size range of 15-28 nm. Se NPs@Cs-sialic acid were stable for approximately 60 days at 4 ℃. The as-synthesized NPs exerted inhibitory effects on T98 greater than 3 T3 > A172 cells in a dose- and time-dependent manner. Additionally, sialic acid ameliorated the blood biocompatibility of Se NPs@Cs. Taken together, sialic acid improved both the stability and biological activity of Se NPs@Cs.


Assuntos
Antineoplásicos , Quitosana , Glioblastoma , Nanopartículas , Selênio , Humanos , Selênio/farmacologia , Selênio/química , Quitosana/química , Ácido N-Acetilneuramínico , Glioblastoma/tratamento farmacológico , Antineoplásicos/farmacologia , Linhagem Celular , Nanopartículas/química
12.
Brain Behav ; 13(5): e2986, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37032465

RESUMO

OBJECTIVES: In the study, we examined the effects of ketamine and extremely low-frequency electromagnetic fields (ELF-EMF) on depression-like behavior, learning and memory, expression of GFAP, caspase-3, p53, BDNF, and NMDA receptor in animals subjected to chronic unpredictable stress (CUS). METHODS: After applying 21 days of chronic unpredictable stress, male rats received intraperitoneal (IP) of ketamine (5 mg/kg) and then were exposed to ELF-EMF (10-Hz, 10-mT exposure conditions) for 3 days (3 h per day) and behavioral assessments were performed 24 h after the treatments. Instantly after the last behavioral test, the brain was extracted for Nissl staining, immunohistochemistry, and real-time PCR analyses. Immunohistochemistry (IHC) was conducted to assess the effect of ketamine and ELF-EMF on the expression of astrocyte marker (glial fibrillary acidic protein, GFAP) in the CA1 area of the hippocampus and medial prefrontal cortex (mPFC). Also, real-time PCR analyses were used to investigate the impacts of the combination of ketamine and ELF-EMF on the expression of caspase3, p53, BDNF, and NMDA receptors in the hippocampus in rats submitted to the CUS procedure. Results were considered statistically significant when p < .05. RESULTS: Our results revealed that the combination of ketamine and ELF-EMF increased depression-like behavior, increased degenerated neurons and decreased the number of GFAP (+) cells in the CA1 area and mPFC, incremented the expression of caspase-3, and reduced the expression of BDNF in the hippocampus but showed no effect on the expression of p53 and NMDA-R. CONCLUSIONS: These results reveal that combining ketamine and ELF-EMF has adverse effects on animals under chronic unpredictable stress (CUS).


Assuntos
Ketamina , Ratos , Masculino , Animais , Ketamina/farmacologia , Caspase 3 , Depressão/etiologia , Campos Eletromagnéticos/efeitos adversos , Fator Neurotrófico Derivado do Encéfalo , Proteína Supressora de Tumor p53
13.
Int J Biol Macromol ; 239: 124099, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-36948335

RESUMO

Wound dressing is applied to promote the healing process, wound protection, and additionally regeneration of injured skin. In this study, a bilayer scaffold composed of a hydrogel and nanofibers was fabricated to improve the regeneration of injured skin. To this end, polycaprolactone/gelatin (PCL/Gel) nanofibers were electrospun directly on the prepared collagen/alginate (Col/Alg) hydrogel. The bilayer scaffold was characterized by scanning electron microscopy (SEM), Fourier transform infrared (FTIR), mechanical properties, and swelling/degradation time. Cytotoxicity assays were evaluated using MTT assay. Then, the nanofiber and bilayer scaffolds were seeded with Adipose-derived stem cells (ADSCs). ADSCs were isolated from rat adipose tissue and analyzed using flow cytometry, in advance. Full-thickness wounds on the backs of rats were dressed with ADSCs-seeded bilayer scaffolds and nanofibers. Histopathological evaluations were performed after 14 and 21 days using H&E (hematoxylin and eosin) staining. The results indicated that re-epithelialization, angiogenesis, and collagen remodeling were enhanced in ADSCs-seeded bilayer scaffolds and nanofibers in comparison with the control group. In conclusion, the best re-epithelialization, collagen organization, neovascularization, and low presence of inflammation in the wound area were observed in the ADSCs-seeded bilayer scaffolds.


Assuntos
Células-Tronco Mesenquimais , Nanofibras , Ratos , Animais , Gelatina , Alicerces Teciduais , Hidrogéis , Alginatos , Colágeno , Bandagens
14.
Sci Rep ; 12(1): 19806, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396965

RESUMO

Dexamethasone, a common medication used in the treatment regimen of glioblastoma, has broad inhibitory effects on the immune responses. Here, in an in vitro study, we examined the effects of piroxicam, a potent substitute for dexamethasone, on peripheral blood mononuclear cells (PBMCs) co-cultured with two glioblastoma cell lines, U-87 MG and A-172 cells. MTT assay was used to determine the proliferation of PBMCs treated with piroxicam, or dexamethasone. In addition, to evaluate the effects of drugs on the cell cycle distribution, DNA content per cell was analyzed in PBMCs and A-172 cell lines using flow cytometry. Oxidative parameters, including superoxide dismutase-3 (SOD3) activity and total anti-antioxidant capacity, lactate dehydrogenase (LDH) activity, as well as IFN-γ and TGF-ß levels were measured in PBMCs alone or in the presence of cell lines using ELISA. Unlike dexamethasone, piroxicam showed a protective effect on PBMCs against both glioblastoma cell lines. Furthermore, while dexamethasone reduced the proliferation of PBMCs, piroxicam had no adverse effect on the proliferation. Cell cycle analysis showed a reduction in the G2/M phase in piroxicam-treated A-172 cells. Additionally, dexamethasone limited the cell cycle progression by increasing the fraction of PBMCs in G0/G1. Interestingly, after co-culturing piroxicam-treated PBMCs with cell lines, a remarkable rise in the LDH activity was observed. Although not significant, piroxicam partially decreased TGF-ß levels in both cell lines. Our findings suggested a protective effect of piroxicam, but not dexamethasone, on PBMCs against inhibitory mechanisms of two glioblastoma cell lines, U-87 and A-172 cells.


Assuntos
Glioblastoma , Leucócitos Mononucleares , Humanos , Leucócitos Mononucleares/metabolismo , Piroxicam/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Linhagem Celular , Fator de Crescimento Transformador beta/metabolismo
15.
Hematol Oncol Stem Cell Ther ; 14(1): 1-15, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32971031

RESUMO

Glioblastoma (GB) is one of the most malignant types of central nervous system tumours, classified as grade IV by the World Health Organization. Despite the therapeutic advances, the prognosis is ominous, with a median survival of about 12-15 months post diagnosis. Although therapeutic options available can increase the survival, they are ineffective in treating patients with GB. Impairing factors such as the blood-brain barrier, cancer stem cells, and infiltration into brain parenchyma lead to failure of current therapies. Therefore, clinicians need novel/alternative effective strategies to treat GB. Due to their ability to preserve healthy tissues and to provide an effective and long-lasting response, stem cells (SCs) with tropism for tumour cells have attracted considerable attention in the scientific community. As is the case here, SCs can be used to target brain tumour cancer cells, especially high-grade malignant gliomas like GB, by overcoming the resistance and exerting benefits for patients affected with such lethal disease. Herein, we will discuss the research knowledge regarding SC-based therapy for the treatment of GB, focalising our attention on SCs and SC-released extracellular vesicles modified to express/load different antitumour payloads, as well as on SCs exploited as a diagnostic tool. Advantages and unresolved issues of anticancer SC-based therapy will also be considered.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Terapia Baseada em Transplante de Células e Tecidos , Glioblastoma , Células-Tronco Neoplásicas , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Prognóstico
16.
Iran J Allergy Asthma Immunol ; 20(3): 314-325, 2021 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-34134453

RESUMO

Perturbed expression of microRNAs (miRs) has been reported in different diseases including autoimmune and chronic inflammatory disorders. In this study, we investigated the expression of miR-25-3p and its targets in the central nervous system (CNS) tissue from mice with experimental autoimmune encephalomyelitis (EAE). We also analyzed the expression of miR-25 and its targets in activated macrophages and splenocytes. EAE was induced in 12-week old female C57BL/6 mice; using myelin oligodendrocyte glycoprotein 35-55/complete Freund's adjuvant (MOG35-55/CFA) protocol. The expression of miR-25-3p and its targets, as well as the expression of inflammatory cytokines, were analyzed. We next established primary macrophage cultures as well as splenocyte cultures and evaluated the levels of miR-25-3p and its target genes in these cells following activation with lipopolysaccharide (LPS) and anti-CD3/anti-CD28 antibodies, respectively. MiR-25-3p expression showed a strong positive correlation with the expression of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1α, and IL-6 pro-inflammatory cytokines. The expression of phosphatase and tensin homolog (Pten) and Krüppel-like factor 4 (Klf4) was significantly reduced at the peak of the disease. Interestingly, Pten and Klf4 expression showed a significant negative correlation with miR-25-3p. Analysis of miR-25-3p expression in LPS-treated primary macrophages revealed significant upregulation in cells treated with 100ng/ml of LPS. This was associated with suppressed levels of miR-25-3p targets in these cells. However, anti-CD3/anti-CD28-stimulated splenocytes failed to show any alterations in miR-25-3p expression compared with vehicle-treated cells. Our results indicate that miR-25-3p expression is likely induced by inflammatory mediators during autoimmune neuroinflammation. This upregulation is associated with decreased levels of Pten and Klf4, genes with known roles in cell cycle regulation and inflammation.


Assuntos
Citocinas/metabolismo , Encefalomielite Autoimune Experimental/enzimologia , Mediadores da Inflamação/metabolismo , Macrófagos/enzimologia , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Baço/enzimologia , Linfócitos T/enzimologia , Animais , Autoimunidade , Células Cultivadas , Citocinas/genética , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Adjuvante de Freund , Regulação da Expressão Gênica , Fator 4 Semelhante a Kruppel/genética , Fator 4 Semelhante a Kruppel/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Glicoproteína Mielina-Oligodendrócito , PTEN Fosfo-Hidrolase/genética , Fragmentos de Peptídeos , Transdução de Sinais , Baço/imunologia , Linfócitos T/imunologia
17.
Brain Res Bull ; 171: 75-83, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33753209

RESUMO

Kindling results in abnormal synaptic potentiation and significant impairment in learning and memory. Electromagnetic field (EMF) effects on learning and memory in kindled animals and its effects on hippocampal neural activity are largely unknown. In the current study, the effects of EMF on learning and memory, as well as hippocampal synaptic plasticity, in kindled rats were investigated. EMF (10 mT; 100 Hz) was applied to fully kindled animals one hour/day for a period of one week. The behavioral and electrophysiological studies were performed 24 h following the EMF application. The kindled rats showed spatial learning deficits during the training phase of the Morris water maze (MWM) test. Moreover, there were increments in escape latency and path length compared to the sham group. The kindled rats spent less time in the target-quadrant probe test, indicating spatial memory impairment. Applying EMF to the KEMF group (kindling + EMF) restored learning and memory, and decreased escape latency and path length significantly compared to the kindled group. EMF alone had no significant effects on the learning and memory parameters. Based on the open field (OF) test results, EMF alone in the EMF group, but not in the kindled or the KEMF groups, decreased the total traveled distance and increased the spent time in the peripheral zone, compared to the sham group. Based on electrophysiological results, applying EMF in the KEMF group returned the ability of synaptic potentiation to the hippocampal CA1 area and high-frequency stimulation induced long-term potentiation (LTP). Accordingly, EMF can be considered a potential therapy for seizure-induced deficits in learning and memory.


Assuntos
Disfunção Cognitiva/fisiopatologia , Campos Eletromagnéticos , Hipocampo/fisiopatologia , Excitação Neurológica/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Cognição/fisiologia , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Ratos , Ratos Wistar , Aprendizagem Espacial/fisiologia
18.
Cancer Biother Radiopharm ; 36(7): 579-587, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32644826

RESUMO

Background: Glioblastoma is a malignant and very aggressive brain tumor with a poor prognosis. Despite having chemotherapy concomitant with surgery and/or radiation therapy, the median survival of glioblastoma-affected people is less than 1 year. Temozolomide (TMZ) is a chemotherapeutic used as a first line treatment of glioblastoma. Several studies have reported that resistance to TMZ due to overexpression of O6-methylguanine-DNA methyltransferase (MGMT) is the main reason for treatment failure. Several studies described that pulsed-electromagnetic field (EMF) exposure could induce cell death and influence gene expression. Materials and Methods: In this study the authors assessed the effects of EMF (50 Hz, 70 G) on cytotoxicity, cell migration, gene expression, and protein levels in TMZ-treated T98 and A172 cell lines. Results: In this study, the authors show that treatment with a combination of TMZ and EMF enhanced cell death and decreased the migration potential of T98 and A172 cells. The authors also observed overexpression of the p53 gene and downregulation of cyclin-D1 protein in comparison to controls. In addition, T98 cells expressed the MGMT protein following treatment, while the A172 cells did not express MGMT. Conclusion: Their data indicate that EMF exposure improved the cytotoxicity of TMZ on T98 and A172 cells and could partially affect resistance to TMZ in T98 cells.


Assuntos
Neoplasias Encefálicas/terapia , Ciclina D2/biossíntese , Metilases de Modificação do DNA/biossíntese , Enzimas Reparadoras do DNA/biossíntese , Glioblastoma/terapia , Magnetoterapia , Temozolomida/farmacologia , Proteína Supressora de Tumor p53/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Ciclina D2/genética , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Campos Eletromagnéticos , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Proteína Supressora de Tumor p53/genética
19.
Sci Rep ; 10(1): 10141, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32576847

RESUMO

Empathy for pain depends on the ability to feel, recognize, comprehend and share painful emotional conditions of others. In this study, we investigated the role of NO in a rat model of empathic pain. Pain was socially transferred from the sibling demonstrator (SD) who experienced five formalin injection to the naïve sibling observer (SO) through observation. SO rats received L-NAME (a nonspecific NO synthase inhibitor) or L-arginine (a precursor of NO) prior to observing the SD. Nociception, and concentrations of NO metabolites (NOx) in the serum, left and right hippocampus, prefrontal cortex, and cerebellum were evaluated. Nociceptive responses were significantly increased in the pain-observing groups. NOx levels measured 24 h after the last pain observation using the Griess method, were indicative of NOx concentration decreases and increases in the left hippocampus and cerebellum, respectively. There was an increase in tissue concentration of NOx in cerebellum and prefrontal cortex in both pain and observer groups 7 days after the fifth formalin injection. Our results suggest that NO is involved in development of empathic hyperalgesia, and observation of sibling's pain can change NO metabolites in different brain regions in observer rats.


Assuntos
Empatia , Hiperalgesia/etiologia , Óxido Nítrico/fisiologia , Nociceptividade/fisiologia , Dor/etiologia , Dor/psicologia , Animais , Arginina/farmacologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Empatia/efeitos dos fármacos , Feminino , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Nociceptividade/efeitos dos fármacos , Ratos Wistar
20.
Daru ; 27(2): 533-539, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31317441

RESUMO

BACKGROUND: Exosomes are natural nanoparticles that are involved in intercellular communication via transferring molecular information between cells. Recently, exosomes have been considered for exploitation as novel drug delivery systems due to their specific properties for carrying specific molecules and surface proteins. METHODS: In this study, U-87 cell derived exosomes have been investigated for delivery of a potent chemotherapeutic agent, paclitaxel (PTX). Two methods of loading were utilized to incorporate PTX in exosomes and the exosomes pharmaceutical and cytotoxic characterizations were determined. RESULTS: The drug loaded and empty exosomes were found to have particle size of 50-100 nm and zeta potential of ≈ - 20 mV. Loading capacity of 7.4 ng and 9.2 ng PTX into 1 µg of exosome total protein were also measured for incubation and sonication methods, respectively. Incorporation of PTX into exosomes significantly increased its cytotoxicity against U-87 cell line (59.92% cell viability) while it was found that the empty exosomes exhibited cell viability of 91.98%. CONCLUSIONS: Loading method could affect the loading capacity of exosomes and their encapsulated chemotherapeutic molecule showed higher cytotoxicity into exosomes. These results promise exosomes as appropriate drug delivery system for glioblastoma multiform (GBM) treatment.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Encefálicas/metabolismo , Exossomos/química , Glioblastoma/metabolismo , Paclitaxel/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos , Exossomos/ultraestrutura , Glioblastoma/tratamento farmacológico , Humanos , Tamanho da Partícula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA