Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Res ; 1828: 148764, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38242524

RESUMO

Mesenchymal stem cells therapy provides a new perspective of therapeutic approaches in the treatment of neurodegenerative diseases. The present study aimed to investigate the effects of intranasally transplanted human "olfactory ecto-mesenchymal stem cells" (OE-MSCs) in Alzheimer's disease (AD) rats. In this study, we isolated OE-MSCs from human olfactory lamina propria and phenotypically characterized them using immunocytochemistry and flow cytometry. The undifferentiated OE-MSCs were transplanted either by intranasal (IN) or intrahippocampal (IH) injection to rat models of AD, which were induced by injecting amyloid-beta (Aß) intrahippocampally. Behavioral, histological, and molecular assessments were performed after a three-month recovery period. Based on the results, intranasal administration of OE-MSCs significantly reduced Aß accumulation and neuronal loss, improved learning and memory impairments, and increased levels of BDNF (brain-derived neurotrophic factor) and NMDAR (N-methyl-D-Aspartate receptors) in the AD rat model. These changes were more significant in animals who received OE-MSCs by intranasal injection. The results of this study suggest that OE-MSCs have the potential to enhance cognitive function in AD, possibly mediated by BDNF and the NMDA receptors.


Assuntos
Doença de Alzheimer , Células-Tronco Mesenquimais , Humanos , Ratos , Animais , Doença de Alzheimer/patologia , Aprendizagem Espacial , Fator Neurotrófico Derivado do Encéfalo , Administração Intranasal , Peptídeos beta-Amiloides , Transtornos da Memória/terapia , Células-Tronco Mesenquimais/fisiologia , Modelos Animais de Doenças
2.
Toxicol Appl Pharmacol ; 481: 116754, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37956929

RESUMO

Glioblastoma multiforme (GBM) is one of the most vascular among solid tumors, and despite the use of multimodal therapies, the survival of these patients is poor. In order to target angiogenesis in GBM as a promising strategy, an antiangiogenic drug is required. This study was designed to evaluate the effects of sunitinib, a multityrosine kinase inhibitor with tumor proliferation and angiogenesis inhibitory properties, on GBM-bearing rats. Given the ineffective drug delivery to the brain due to the presence of the blood-brain barrier (BBB), intra-nasal (IN) drug delivery has recently been considered as a non-invasive method to bypass BBB. Therefore, in the current study, IN was used as an ideal method for the delivery of sunitinib to the brain, and the effects of this method were also compared to the OR administration of the sunitinib. GBM was induced in the brain of male Wistar rats, and they were randomly divided into 4 groups; IN-STB (sunitinib intranasal delivery), IN-sham (placebo intranasal delivery), OR-STB (sunitinib oral delivery) and OR-sham (placebo oral delivery). After the end of the treatment period, an MRI of animals' brains showed a reduction in tumor growth in the treatment groups. Immunohistochemistry revealed that sunitinib inhibits angiogenesis in GBM in both OR and IN delivery methods. Analysis of liver tissue and enzymes showed that IN delivery of sunitinib had less hepatotoxicity than the OR method. Overall, it was found that IN sunitinib delivery could be used as a potential non-hepatotoxic alternative for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Humanos , Masculino , Ratos , Angiogênese , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Ratos Wistar , Sunitinibe/uso terapêutico
3.
Basic Clin Neurosci ; 13(1): 57-70, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36589026

RESUMO

Introduction: Cell therapy is the most advanced treatment of peripheral nerve injury. This study aimed to determine the effects of transplantation of hair follicle stem cells on the regeneration of the sciatic nerve injury in rats. Methods: The bulge region of the rat whisker were isolated and cultured. Morphological and biological features of the cultured bulge cells were observed by light microscopy and immunocytochemistry methods. Percentages of CD34, K15, and nestin cell markers expression were demonstrated by flow cytometry. Rats were randomly divided into 3 groups of injury, epineurium, and epineurium with cells in which rat Hair Follicular Stem Cells (rHFSCs) were injected into the site of the nerve cut. HFSCs were labeled with Bromodeoxyuridine (BrdU), and double-labeling immunofluorescence was performed to study the survival and differentiation of the grafted cells. After 8 weeks, electrophysiological, histological, and immunocytochemical analysis assessments were performed. Results: Rat hair follicle stem cells are suitable for cell culture, proliferation, and differentiation. The results suggest that transplantation of rat hair follicle stem cells can regenerate sciatic nerve injury; moreover, electrophysiology and histology examinations show that sciatic nerve repair was more effective in the epineurium with cell group than in the other experimental group (P<0.05). Conclusion: The achieved results propose that hair follicle stem cells improve axonal growth and functional recovery after peripheral nerve injury. Highlights: This study showed that rat hair follicle stem cells are suitable for cell culture, proliferation and differentiationThe results suggested that transplantation of rat hair follicle stem cells had the potential capability of regenerating sciatic nerve injuryEvidence of electrophysiology and histology showed Concomitant use of epineurium with hair follicle stem cell was more effective repairment. Plain Language Summary: Although repairing damaged peripheral nerves has always been a medical challenge, but peripheral nerve injury has been successfully repaired using various procedures such as nerve auto-graft or stem cell therapy. The functional reconstruction is the most important after therapy because of that primary nerve repair or use of nerve autograft, are still accepted as golden standard methods for treatment. Considerable recent interest has been focused on adult stem cells for both research and clinical applications. A highly promising source of relatively abundant and accessible, active, multipotent adult stem cells are obtained from hair follicles. In research the hair follicle stem cells implanted into the gap region of a severed sciatic nerve injury greatly enhanced the rate of nerve regeneration and the restoration of nerve function. Time is one of the several aspects require specific attention in the clinical treatment of peripheral nerve injury. Because delay of nerve injury treatment may cause neurobiological alterations in neurons and Schwann cells, impairing nerve functional recovery and affect neuron survival. In this study, concluded that stem cell injection 2 weeks after injury in the damaged nerve epineurium repairs nerve fibers, while electrophysiology of the leg muscles showed that muscle function was significantly improved. It indicates the repair of muscular innervation and nerve repair. The results pave the way for further research on this topic.

4.
Life Sci ; 282: 119823, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34273375

RESUMO

AIMS: Zinc oxide nanoparticles (ZnO-NPs) are currently applied in food and pharmaceutical industries whose neurotoxic effect on the central nervous system (CNS) is a major concern. Considering the pharmacological properties (antioxidant, anti-inflammatory) of the geraniol (GE), we aimed to investigate the efficacy of geraniol on ZnO-NPs neurotoxicity. MATERIALS AND METHODS: We used 32 male Wistar rats, randomly assigned to four groups (n = 8): Control, GE (daily received 100 mg/kg of GE by gavage), ZnO-NPs (received intraperitoneal injection of 75 mg/kg of ZnO-NPs twice a week), and ZnO-NPs + GE (received both GE and ZnO-NPs at same doses above during 4 weeks). Morris water maze (MWM) and Y-maze tasks were done to evaluate learning and memory function. Biochemical assays were done to measure total antioxidant capacity (TAC), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPX) and ZnO-NPs bioaccumulation. Nissl and H&E staining were performed for histological evaluations. KEY FINDINGS: The results of behavioral study revealed that GE improved learning and memory impairment induced by ZnO-NPs. Moreover, neuroprotective effect of GE significantly decreased pathological parameters such as necrosis and gliosis, and consequently increased the number of nerve cells in the cortex and different hippocampal areas. Furthermore, biochemical studies demonstrated that GE significantly increased antioxidant indices (namely, TAC, SOD, and GPX) and reduced oxidative stress marker (MDA) and Zn bioaccumulation in ZnO-NPs treated animals. SIGNIFICANCE: Our results provide experimental evidence to further investigate the precise mechanisms underlying the geraniol as a promising therapeutic approach for improvement of cognitive function and neurotoxicity induce by ZnO-NPs.


Assuntos
Monoterpenos Acíclicos/farmacologia , Antioxidantes/farmacologia , Transtornos da Memória , Síndromes Neurotóxicas , Estresse Oxidativo/efeitos dos fármacos , Óxido de Zinco/toxicidade , Animais , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/fisiopatologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/fisiopatologia , Ratos , Ratos Wistar
5.
J Photochem Photobiol B ; 221: 112207, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34119804

RESUMO

Transcranial photobiomodulation (tPBM) is the process of delivering light photons through the skull to benefit from its modifying effect. Brain disorders are important health problems. The aim of this review was to determine the existing evidence of effectiveness, useful parameters, and safety of tPBM in the management of traumatic brain injury, stroke, Parkinson, and Alzheimer's disease as the common brain disorders. Four online databases, including Cochrane, Pub Med, Embase, and Google scholar were searched according to the Preferred Reporting Items for Systematic Reviews and meta-analyses (PRISMA) guidelines. 4728 articles were obtained in the initial search. Only those articles that were published until September 2020 and designed as randomized clinical trials (RCTs) or animal-controlled studies were included. 6 RCTs, 2 related supplementary articles, and 38 controlled animal studies met the inclusion criteria of this study. No RCTs were performed in the fields of Alzheimer's and Parkinson's diseases. The human RCTs and animal studies reported no adverse events resulted from the use of tPBM. Useful parameters of tPBM were identified according to the controlled animal studies. Since the investigated RCTs had no homogenous results, making an evidence-based decision for definite therapeutic application of tPBM is still unattainable. Altogether, these data support the need for large confirmatory well-designed RCTs for using tPBM as a novel, safe, and easy-to-administer treatment of brain disorders. EVIDENCE BEFORE THIS STUDY: High prevalence and complications of brain disorders and also side effects of neuropsychiatric medications have encouraged researchers to find alternative therapeutic techniques which tPBM can be one of them. In present review we tried to determine the existing evidence of effectiveness, useful parameters, and safety of tPBM in the management of traumatic brain injury, stroke, Alzheimer, and Parkinson's disease as common brain disorders. Four online databases, including "Cochrane", "Pub Med", "Embase", and "Google scholar" were searched. Only those articles that were published until September 2020 and designed as RCTs or animal-controlled studies were included. Search keywords were the followings: transcranial photobiomodulation" OR "transcranial low-level laser therapy" AND "stroke" OR "traumatic brain injury" OR "Alzheimer" OR "Parkinson". Several studies have confirmed effectiveness of tPBM in treatment of different brain disorders but the level of evidence of its effectiveness remain to be determined. ADDED VALUE OF THIS STUDY: In this study we systematically reviewed human RCTs to determine the existing evidence of tPBM effectiveness in management of four mentioned brain disorders. Since the outcomes of the reviewed RCTs were not homogeneous, further well-designed RCTs are required to decide more definitively on the evidence of this noninvasive and probably safe therapeutic intervention. We hypothesized that non-homogeneous outcomes could be due to inefficiency of PBM parameters. Controlled animal studies have the advantage of using objective tests to evaluate the results and compare them with the control group. We determined useful tPBM parameters based on these studies. IMPLICATIONS OF ALL THE AVAILABLE EVIDENCE: This research is part of our main project of tinnitus treatment using photobiomodulation (PBM). Evidence of central nervous system involvement in tinnitus led us to believe that treatment protocol of tinnitus should also include transcranial PBM. The determined useful parameters can be helpful in designing more efficient tPBM protocols in the management of brain disorders and tinnitus as a common debilitating symptom that can be associated with these disorders.


Assuntos
Encefalopatias/radioterapia , Terapia com Luz de Baixa Intensidade , Doença de Alzheimer/radioterapia , Animais , Lesões Encefálicas Traumáticas/radioterapia , Modelos Animais de Doenças , Humanos , Doença de Parkinson/radioterapia , Ensaios Clínicos Controlados Aleatórios como Assunto
6.
Neurosci Lett ; 746: 135652, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33482310

RESUMO

Methamphetamine (METH) may cause long‒lasting neurotoxic effects and cognitive impairment. On the other hand, the ovarian hormones estrogen and progesterone have neuroprotective effects. In the current study, we aimed to examine the effects of estrogen and progesterone on anxiety‒like behavior and neuronal damage in METH‒exposed ovariectomized (OVX) rats. Three weeks after ovariectomy, the animals received estrogen (1 mg/kg, i.p.), or progesterone (8 mg/kg, i.p.), or estrogen plus progesterone (with the same doses), or vehicle during 7 consecutive days (days 22-28). On day 28, OVX rats were exposed to a single‒day METH regimen (6 mg/kg, four s.c. Injections, with 2 h interval) 30 min after the hormone treatment. The next day (on day 29), the animals were assessed for anxiety‒related behaviors using the open field and elevated plus‒maze tasks. The animals were then sacrificed and brain water content, cell apoptosis and expression of IL-1ß were evaluated. The findings showed that treatment with estrogen or progesterone alone in METH‒exposed rats significantly improved hyperthermia, anxiety‒like behavior, neuronal damage, and inflammation in the CA1 area. Also, treatment with estrogen plus progesterone improved hyperthermia and brain edema. Taken together, the findings suggest that treatment with ovarian hormones can partially prevent hyperthermia and anxiety‒related behaviors induced by METH in OVX rats, which could be accompanied by their neuroprotective effects in the hippocampus.


Assuntos
Ansiedade/metabolismo , Encéfalo/metabolismo , Estrogênios/uso terapêutico , Metanfetamina/toxicidade , Ovariectomia/efeitos adversos , Progesterona/uso terapêutico , Animais , Ansiedade/induzido quimicamente , Ansiedade/prevenção & controle , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Estimulantes do Sistema Nervoso Central/toxicidade , Estrogênios/farmacologia , Feminino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ovário/metabolismo , Progesterona/farmacologia , Ratos , Ratos Wistar
7.
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
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.
Biomed Pharmacother ; 92: 254-264, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28551545

RESUMO

Glioblastoma multiforme (GBM) is the most malignant brain cancer that causes high mortality in humans. It responds poorly to the most common cancer treatments, such as surgery, chemo- and radiation therapy. Temozolomide (TMZ) is an alkylating agent that has been widely used to treat GBM; 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. In this study, we evaluated the potential synergistic effect of 100µM TMZ and EMF (100Hz, 100G) on two human glioma cells line, i.e., U87 and T98G above single treatments, TMZ or EMF. Co-treatment synergistically enhanced apoptosis in U87 and T98G cells, by increasing the expression of P53, Bax, and Caspase-3 and decreasing that of Bcl-2 and Cyclin-D1. We also observed an increase in reactive oxygen species (ROS) production and the overexpression of the heme oxygenase-1 (HO-1) gene in comparison to controls. In conclusion, since EMF enhanced the apoptotic effect of TMZ, possibly through a redox regulation mechanism, the TMZ/EMF combination may be effective for glioma cancer treating. Further studies are needed to reveal the action mechanism of this possible novel therapeutic approach.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Citotoxinas/toxicidade , Dacarbazina/análogos & derivados , Campos Eletromagnéticos/efeitos adversos , Glioblastoma/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Dacarbazina/toxicidade , Glioblastoma/patologia , Humanos , Magnetoterapia/métodos , Temozolomida
10.
Electromagn Biol Med ; 36(3): 238-247, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27874284

RESUMO

The impact of extremely low-frequency pulsed electromagnetic fields (ELF-PEMFs) at various frequencies and amplitudes was investigated on cell cycle, apoptosis and viability of the Glioblastoma Multiforme (GBM) cell line (U87), in vitro. The GBM is a malignant brain tumor with high mortality in humans and poorly responsive to the most common type of cancer treatments, such as surgery, chemotherapy and radiation therapy. U87 cells with five experimental groups (I-V) were exposed to various ELF-PEMFs for 2, 4 and 24 h, as follows: (I) no exposure, control; (II) 50 Hz 100 ± 15 G; (III) 100 Hz 100 ± 15 G; (IV) 10 Hz 50 ± 10 G; (V) 50 Hz 50 ± 10 G. The morphology properties, cell viability and gene expression of proteins involved in cell cycle regulation (Cyclin-D1 and P53) and apoptosis (Caspase-3) were investigated. After 24 h, the cell viability and Cyclin-D1 expression increased in Group II (30%, 45%), whereas they decreased in Groups III (29%, 31%) and IV (21%, 34%); P53 and Caspase-3 elevated only in Group III; and no significant difference was observed in Group V, respectively, compared with the control (p < 0.05). The data suggest that the proliferation and apoptosis of human GBM are influenced by exposure to ELF-PEMFs in different time-dependent frequencies and amplitudes. The fact that some of the ELF-PEMFs frequencies and amplitudes favor U87 cells proliferation indicates precaution for the use of medical devices related to the MFs on cancer patients. On the other hand, some other ELF-PEMFs frequencies and intensities arresting U87 cells growth could open the way to develop novel therapeutic approaches.


Assuntos
Campos Eletromagnéticos , Glioblastoma , Apoptose , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclina D1/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA