Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Pharm ; 20(11): 5921-5936, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37874541

RESUMO

Prostate cancer (PCa) is the most prevalent cause of cancer deaths in men. Conventional strategies, such as surgery, radiation, or chemotherapy, face challenges including poor prognosis and resistance. Therefore, the development of new improved strategies is vital to enhance patient outcomes. Recently, immunotherapy has shown potential in the treatment of a range of cancers, including PCa. Tumor-associated macrophages (TAMs) play an important role in the tumor microenvironment (TME) and reprogramming of TAMs is associated with remodeling the TME. The colony-stimulating factor-1/colony stimulating factor-1 receptor (CSF-1/CSF-1R) signaling pathway is closely related to the polarization of TAMs. The downregulation of CSF-1R, using small interfering RNA (siRNA), has been shown to achieve the reprogramming of TAMs, from the immunosuppressive M2 phenotype to the immunostimulatory M1 one. To maximize specific cellular delivery an M2 macrophage-targeting peptide, M2pep, was formulated with an amphiphilic cationic ß-Cyclodextrin (CD) incorporating CSF-1R siRNA. The resulting nanoparticles (NPs) increased M2 macrophage targeting both in vitro and in vivo, promoting the release of M1 factors and simultaneously downregulating the levels of M2 factors through TAM reprogramming. The subsequent remodeling of the TME resulted in a reduction in tumor growth in a subcutaneous PCa mouse model mainly mediated through the recruitment of cytotoxic T cells. In summary, this M2pep-targeted CD-based delivery system demonstrated significant antitumor efficacy, thus presenting an alternative immunotherapeutic strategy for PCa treatment.


Assuntos
Ciclodextrinas , Nanopartículas , Neoplasias da Próstata , Masculino , Camundongos , Animais , Humanos , RNA Interferente Pequeno , Microambiente Tumoral , Neoplasias da Próstata/terapia , Neoplasias da Próstata/tratamento farmacológico , Imunoterapia/métodos
2.
Eur J Pharm Sci ; 185: 106427, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-36948408

RESUMO

Prostate cancer remains a serious condition threatening the health of men. Due to the complicated nature of the tumour microenvironment (TME), conventional treatments face challenges including poor prognosis and tumour resistance, therefore new therapeutic strategies are urgently needed. Small interfering RNA (siRNA), a double-stranded non-coding RNA, regulates specific gene expression through RNA interference. Tumour-associated macrophages (TAMs) are a potential therapeutic target in cancer immunotherapy. Colony stimulating factor-1/colony stimulating factor-1 receptor (CSF-1/CSF-1R) signaling pathway plays a crucial role in the polarization of the immunosuppressive TAMs, M2 macrophages. Downregulation of CSF-1R is known to reprogram the immunosuppressive TAMs, M2 macrophages, to the immunostimulatory phenotype, M1 macrophages. Sialic acid is a ligand for Siglec-1 (CD169) which is overexpressed on M2 macrophages with little expression in other phenotypes. Therefore, a sialic acid-targeted cyclodextrin-based nanoparticle was developed to specifically deliver CSF-1R siRNA to M2 macrophages. The nanoparticles were studied in vitro using both human and mouse prostate cancer cell lines. Results show that the targeted nanoparticles achieved cell specific delivery to M2 macrophages via the sialic acid-CD169 axis. The expression of CSF-1R was significantly downregulated in M2 macrophages (29.64% for targeted vs 19.31% for non-targeted nanoparticles in THP-1-derived M2 macrophages and 38.94% for targeted vs 18.51% for non-targeted nanoparticles in RAW 264.7-derived M2 macrophages, n = 4, p < 0.01). The resulting reprograming of M2 macrophages to M1 enhanced the level of apoptosis in the prostate cancer cells in a Transwell model (49.17% for targeted vs 37.68% for non-targeted nanoparticles in PC-3 cells and 69.15% for targeted vs 44.73% for non-targeted nanoparticles in TRAMP C1 cells, n = 3, p < 0.01). Thus, this targeted cyclodextrin-based siRNA drug delivery system provides a potential strategy for prostate cancer immunotherapy.


Assuntos
Ciclodextrinas , Nanopartículas , Neoplasias da Próstata , Animais , Humanos , Masculino , Camundongos , Fatores Estimuladores de Colônias , Imunoterapia/métodos , Ácido N-Acetilneuramínico , Nanopartículas/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Microambiente Tumoral , Macrófagos Associados a Tumor , Receptor de Fator Estimulador de Colônias de Macrófagos/genética
3.
Pharmaceutics ; 15(2)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36839842

RESUMO

Huntington's disease (HD) is a progressive inherited neurodegenerative disease caused by a CAG repeat expansion in the huntingtin gene, which is translated into the pathologic mutant huntingtin (mHTT) protein. Despite the great potential of HTT lowering strategies and the numerous antisense oligonucleotides (ASOs) in pre- and clinical trials, sustained silencing of mHTT has not been achieved. As a strategy to improve ASO delivery, cyclodextrin-based nanoparticles (CDs) offer a promising approach. Here, three CDs with distinct chemical structures were designed and their efficacies were compared as potential platforms for the delivery of ASO targeting HTT. Results using striatal neurons and HD patient-derived fibroblasts indicate that modified γ-CDs exhibited the best uptake efficiency and successfully downregulated mHTT at protein and allele levels. The incorporation of the brain-targeting peptide RVG into the modified γ-CDs showed greater downregulation of mHTT protein and HD-causing allele SNP1 than untargeted ones in an in vitro blood-brain barrier model. Although the ASO sequence was designed as a nonallele-specific therapeutic approach, our strategy gives an additional benefit of some mHTT selectivity. Overall, this study demonstrated the CD platform's feasibility for delivering ASO-based therapeutics for HD treatment.

4.
Drug Discov Today ; 28(3): 103505, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36708760

RESUMO

The successful development of nonviral delivery systems for nucleic acids has been reported extensively over the past number of years. Among them, lipid-based nanoparticles (LNPs) represent the most advanced platform. This review provides an overview of the state-of-the-art in LNP technology, focusing on the delivery of a range of nucleic acids. Recent advances in the development of an efficient and safe lipid-based system are critically analyzed with a particular emphasis on the rationale behind the design of LNPs and on attempts to elucidate the resulting molecular assembly and structure, their interactions with cellular proteins and biodistribution. In addition, manufacturing methods including microfluidics and their potential to influence stability and scale-up are summarized.


Assuntos
Nanopartículas , Ácidos Nucleicos , Distribuição Tecidual , Lipídeos/química , Nanopartículas/química , Microfluídica , RNA Interferente Pequeno
5.
Int J Mol Sci ; 23(17)2022 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-36077202

RESUMO

Non-viral delivery of therapeutic nucleic acids (NA), including siRNA, has potential in the treatment of diseases with high unmet clinical needs such as acute myeloid leukaemia (AML). While cationic biomaterials are frequently used to complex the nucleic acids into nanoparticles, attenuation of charge density is desirable to decrease in vivo toxicity. Here, an anionic amphiphilic CD was synthesised and the structure was confirmed by Fourier-transform infrared spectroscopy (FT-IR), Nuclear Magnetic Resonance (NMR), and high-resolution mass spectrometry (HRMS). A cationic amphiphilic cyclodextrin (CD) was initially used to complex the siRNA and then co-formulated with the anionic amphiphilic CD. Characterisation of the co-formulated NPs indicated a significant reduction in charge from 34 ± 7 mV to 24 ± 6 mV (p < 0.05) and polydispersity index 0.46 ± 0.1 to 0.16 ± 0.04 (p < 0.05), compared to the cationic CD NPs. Size was similar, 161−164 nm, for both formulations. FACS and confocal microscopy, using AML cells (HL-60), indicated a similar level of cellular uptake (60% after 6 h) followed by endosomal escape. The nano co-formulation significantly reduced the charge while maintaining gene silencing (21%). Results indicate that blending of anionic and cationic amphiphilic CDs can produce bespoke NPs with optimised physicochemical properties and potential for enhanced in vivo performance in cancer treatment.


Assuntos
Ciclodextrinas , Leucemia Mieloide Aguda , Nanopartículas , Ânions , Cátions , Ciclodextrinas/química , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Nanopartículas/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Espectroscopia de Infravermelho com Transformada de Fourier
6.
Eur J Pharm Biopharm ; 169: 309-318, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34793942

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by a mutation in the huntingtin (HTT) gene, leading to a toxic version of the HTT protein. There are currently no disease-modifying therapies available. In this scenario, gene-based treatments for HD aimed at lowering HTT levels have become one of the most promising emerging therapeutic options. To date, however, promising results have only been achieved following direct intrathecal or intracranial injections designed to circumvent the blood-brain barrier (BBB). Consequently, efforts to develop less invasive delivery platforms are highly desirable. Here, we described a novel delivery system based on modified cyclodextrin nanoparticles (CDs) loaded with small interfering RNAs (siRNAs) targeting HTT andcomplexed with the rabies virus glycoprotein(RVG), a BBB-shuttle peptide. Results using an in vitro BBB model, indicate the formulation successfully crosses the brain endothelial cells, releases the encapsulated siRNAs into the cytoplasm of neuronal cells, and mediates downregulation of HTT. In conclusion, the CD platform is a promising option for delivery of siRNA-based therapeutics for HD with wider potential to treat other diseases with a genetically validated target in the central nervous system.


Assuntos
Proteína Huntingtina/genética , Doença de Huntington/genética , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Ciclodextrinas/química , Regulação para Baixo/genética , Células Endoteliais/metabolismo , Inativação Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glicoproteínas/química , Humanos , Fragmentos de Peptídeos/química , Ratos , Proteínas Virais/química
7.
Mol Pharm ; 18(4): 1491-1506, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33734715

RESUMO

RNA-based therapeutics have emerged as one of the most powerful therapeutic options used for the modulation of gene/protein expression and gene editing with the potential to treat neurodegenerative diseases. However, the delivery of nucleic acids to the central nervous system (CNS), in particular by the systemic route, remains a major hurdle. This review will focus on the strategies for systemic delivery of therapeutic nucleic acids designed to overcome these barriers. Pathways and mechanisms of transport across the blood-brain barrier which could be exploited for delivery are described, focusing in particular on smaller nucleic acids including antisense oligonucleotides (ASOs) and small interfering RNA (siRNA). Approaches used to enhance delivery including chemical modifications, nanocarrier systems, and target selection (cell-specific delivery) are critically analyzed. Learnings achieved from a comparison of the successes and failures reported for CNS delivery of ASOs versus siRNA will help identify opportunities for a wider range of nucleic acids and accelerate the clinical translation of these innovative therapies.


Assuntos
Doenças do Sistema Nervoso Central/terapia , Portadores de Fármacos/química , Terapia Genética/métodos , Oligonucleotídeos Antissenso/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Doenças do Sistema Nervoso Central/genética , Modelos Animais de Doenças , Composição de Medicamentos , Humanos , Nanopartículas/química , Oligonucleotídeos Antissenso/farmacocinética , Permeabilidade , RNA Interferente Pequeno/farmacocinética
8.
Environ Pollut ; 256: 113484, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31677872

RESUMO

The widespread production and use of silver nanomaterials (AgNMs) in consumer and medical products have been raising environmental concerns. Once in the environment, the soil is one of the major sinks of AgNMs due to e.g. sewage sludge applications, and invertebrates are directly exposed. In this study, we investigate the potential of N-acetylcysteine (NAC) to reduce the toxic effects of Ag NM300 K (and AgNO3) on the soil invertebrate Enchytraeus crypticus. Ag NM300 K induces mortality, reproduction impairment, and avoidance. The addition of NAC to the soil showed a remarkable reduction in the toxicity of Ag, indicating that NAC can act as a detoxifying agent for terrestrial organisms exposed to Ag materials. That the reduction in toxicity likely is caused by thiol groups, was confirmed by GSH and GSSH studies. Identifying the mechanisms and hence alternatives that allow the recovery of contaminated soils is an important mitigation measure to promote environmental safety and reduce the associated risks to human health. Further, it may inform on strategies to implement in safe-by-design industry development.


Assuntos
Nanoestruturas/toxicidade , Oligoquetos/fisiologia , Prata/toxicidade , Poluentes do Solo/toxicidade , Acetilcisteína/metabolismo , Animais , Oligoquetos/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Esgotos , Nitrato de Prata/toxicidade , Solo , Poluentes do Solo/análise
9.
Nanomaterials (Basel) ; 9(6)2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-31195669

RESUMO

Graphene-based nanomaterials (GBNs) possess unique physicochemical properties, allowing a wide range of applications in physical, chemical, and biomedical fields. Although GBNs are broadly used, information about their adverse effects on ecosystem health, especially in the terrestrial environment, is limited. Therefore, this study aims to assess the toxicity of two commonly used derivatives of GBNs, graphene oxide (GO) and reduced graphene oxide (rGO), in the soil invertebrate Enchytraeus crypticus using a reduced full life cycle test. At higher exposure concentrations, GO induced high mortality and severe impairment in the reproduction rate, while rGO showed little adverse effect up to 1000 mg/kg. Collectively, our body of results suggests that the degree of oxidation of GO correlates with their toxic effects on E. crypticus, which argues against generalization on GBNs ecotoxicity. Identifying the key factors affecting the toxicity of GBNs, including ecotoxicity, is urgent for the design of safe GBNs for commercial purposes.

10.
ACS Appl Mater Interfaces ; 10(26): 21891-21900, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29889487

RESUMO

A new nanohydrogel drug delivery platform based on Laponite nanodiscs, polyacrylate, and sodium phosphate salts is described. The hybrid nanohydrogel is tailored to obtain soft and flexible nanohydrogels with G' around 3 kPa, which has been proposed as the ideal stiffness for drug delivery applications. In vitro studies demonstrate that the new nanohydrogels are biocompatible, biodegradable, nonswellable, pH-responsive, and noncytotoxic and are able to deliver antineoplastic drugs into cancer cells. The IC50 of nanohydrogels containing cisplatin, 4-fluorouracil, and cyclophosphamide is significantly lower than the IC50 of the free drugs. In vivo experiments suggest that the new nanomaterials are biocompatible and do not accumulate in crucial organs. The simple formulation procedure enables encapsulation of virtually any water-soluble molecule, without the need for chemical modification of the guests. These nanohydrogels are a versatile platform that enables the simultaneous encapsulation of several cancer drugs, yielding an efficient drug cocktail delivery system, which for instance presents a positive synergistic effect against MCF-7 cells.


Assuntos
Nanoestruturas , Antineoplásicos , Sistemas de Liberação de Medicamentos , Hidrogéis , Silicatos , Nanomedicina Teranóstica
11.
Mol Nutr Food Res ; 61(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28544198

RESUMO

The hyperphosphorylation of microtubule-associated protein tau (tau) in the hippocampus can be caused by central and peripheral insulin resistance and these alterations are related to the development of tauopathies, such as Alzheimer's disease. In this study, we used a high-fat diet to induce obesity and insulin resistance in adult Swiss mice and checked whether supplementation with Myrciaria jaboticaba berry peel for 10 weeks could improve insulin sensitivity, learning/memory performance, and prevent tau phosphorylation in the hippocampus. Furthermore, adipocytokines, inflammatory markers, and oxidative stress were assessed. Myrciaria jaboticaba peel has phenolic compounds (e.g., cyanidin, ellagic acid), dietary fiber and carotenoids, which contribute to great antioxidant capacity. Supplementation of the high-fat diet with 4% M. jaboticaba peel prevented fat weight gain and reduced peripheral insulin resistance. The treated group also showed lower tau phosphorylation in the hippocampus corroborating better learning/memory performance in the Morris water maze test. Maintenance of neuronal viability, lower levels of hippocampal inflammatory markers, and improved brain antioxidant defenses were also related to the consumption of M. jaboticaba peel. These findings contribute to a better understanding of how a high-fat diet supplemented with jaboticaba berry peel counteracts the impairment of cognitive functions caused by high-fat diet intake and diet-induced insulin resistance.


Assuntos
Frutas/química , Resistência à Insulina , Myrtaceae/química , Proteínas tau/metabolismo , Animais , Antocianinas/farmacologia , Biomarcadores/sangue , Carotenoides/farmacologia , Cognição/efeitos dos fármacos , Citocinas/sangue , Dieta Hiperlipídica , Fibras na Dieta/farmacologia , Ácido Elágico/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Insulina/sangue , Masculino , Memória/efeitos dos fármacos , Camundongos , Obesidade/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Aumento de Peso , Proteínas tau/genética
12.
Int J Mol Sci ; 17(11)2016 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-27886057

RESUMO

We have previously demonstrated that Phoneutria nigriventer venom (PNV) causes blood-brain barrier (BBB) breakdown, swelling of astrocytes end-feet and fluid permeation into brain interstitium in rats. Caveolae and water channels respond to BBB alterations by co-participation in shear stress response and edema formation/resolution. Herein, we showed post-natal developmental-related changes of two BBB-associated transporter proteins: the endothelial caveolin-1 (Cav-1), the major scaffolding protein from caveolae frame, and the astroglial aquaporin-4 (AQP4), the main water channel protein expressed in astrocytic peri-vascular end-feet processes, in the hippocampus of rats intraperitoneally-administered PNV. Western blotting protein levels; immunohistochemistry (IHC) protein distribution in CA1, CA2, and CA3 subfields; and gene expression by Real Time-Polymerase Chain Reaction (qPCR) were assessed in post-natal Day 14 (P14) and 8-10-week-old rats over critical periods of envenomation. The intensity and duration of the toxic manifestations indicate P14 neonate rats more vulnerable to PNV than adults. Histologically, the capillaries of P14 and 8-10-week-old rats treated with PNV showed perivascular edema, while controls did not. The intensity of the toxic manifestations in P14 decreases temporally (2 > 5 > 24 h), while inversely the expression of AQP4 and Cav-1 peaked at 24 h when clinically PNV-treated animals do not differ from saline controls. IHC of AQP4 revealed that hippocampal CA1 showed the least expression at 2 h when toxic manifestation was maximal. Subfield IHC quantification revealed that in P14 rats Cav-1 peaked at 24 h when toxic manifestations were absent, whereas in 8-10-week-old rats Cav-1 peaked at 2 h when toxic signs were highest, and progressively attenuated such increases until 24 h, remaining though significantly above baseline. Considering astrocyte-endothelial physical and functional interactions, we hypothesize that age-related modulations of AQP4 and Cav-1 might be linked both to changes in functional properties of astrocytes during post-natal development and in the BBB breakdown induced by the venom of P. nigriventer.


Assuntos
Aquaporina 4/genética , Caveolina 1/genética , Edema/genética , Hipocampo/efeitos dos fármacos , Picada de Aranha/genética , Venenos de Aranha/toxicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Aquaporina 4/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Caveolina 1/metabolismo , Edema/induzido quimicamente , Edema/metabolismo , Edema/patologia , Regulação da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Injeções Intraperitoneais , Masculino , Ratos , Ratos Wistar , Transdução de Sinais , Picada de Aranha/induzido quimicamente , Picada de Aranha/metabolismo , Picada de Aranha/patologia , Venenos de Aranha/administração & dosagem , Venenos de Aranha/isolamento & purificação , Aranhas/química , Aranhas/patogenicidade , Água/metabolismo
13.
Biomed Res Int ; 2014: 721968, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25247186

RESUMO

Phoneutria nigriventer spider accidental envenomation provokes neurotoxic manifestations, which when critical, results in epileptic-like episodes. In rats, P. nigriventer venom (PNV) causes blood-brain barrier breakdown (BBBb). The PNV-induced excitotoxicity results from disturbances on Na(+), K(+) and Ca(2+) channels and glutamate handling. The vascular endothelial growth factor (VEGF), beyond its angiogenic effect, also, interferes on synaptic physiology by affecting the same ion channels and protects neurons from excitotoxicity. However, it is unknown whether VEGF expression is altered following PNV envenomation. We found that adult and neonates rats injected with PNV showed immediate neurotoxic manifestations which paralleled with endothelial occludin, ß-catenin, and laminin downregulation indicative of BBBb. In neonate rats, VEGF, VEGF mRNA, and Flt-1 receptors, glutamate decarboxylase, and calbindin-D28k increased in Purkinje neurons, while, in adult rats, the BBBb paralleled with VEGF mRNA, Flk-1, and calbindin-D28k increases and Flt-1 decreases. Statistically, the variable age had a role in such differences, which might be due to age-related unequal maturation of blood-brain barrier (BBB) and thus differential cross-signaling among components of the glial neurovascular unit. The concurrent increases in the VEGF/Flt-1/Flk-1 system in the cerebellar neuron cells and the BBBb following PNV exposure might imply a cytokine modulation of neuronal excitability consequent to homeostatic perturbations induced by ion channels-acting PNV neuropeptides. Whether such modulation represents neuroprotection needs further investigation.


Assuntos
Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Venenos de Aranha/toxicidade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/efeitos dos fármacos , Masculino , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Aranhas , Regulação para Cima/efeitos dos fármacos
14.
Toxins (Basel) ; 5(12): 2572-88, 2013 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-24351717

RESUMO

Apart from its angiogenic and vascular permeation activity, the vascular endothelial growth factor (VEGF) has been also reported as a potent neuronal protector. Newborn rats with low VEGF levels develop neuron degeneration, while high levels induce protective mechanisms in several neuropathological conditions. Phoneutria nigriventer spider venom (PNV) disrupts the blood-brain barrier (BBB) and causes neuroinflammation in central neurons along with excitotoxic signals in rats and humans. All these changes are transient. Herein, we examined the expression of VEGF and its receptors, Flt-1 and Flk-1 in the hippocampal neurons following envenomation by PNV. Adult and neonatal rats were evaluated at time limits of 2, 5 and 24 h. Additionally, BBB integrity was assessed by measuring the expression of occludin, ß-catenin and laminin and neuron viability was evaluated by NeuN expression. VEGF, Flt-1 and Flk-1 levels increased in PNV-administered rats, concurrently with respective mRNAs. Flt-1 and Flk-1 immunolabeling was nuclear in neurons of hippocampal regions, instead of the VEGF membrane-bound typical location. These changes occurred simultaneously with the transient decreases in BBB-associated proteins and NeuN positivity. Adult rats showed more prominent expressional increases of the VEGF/Flt-1/Flk-1 system and earlier recovery of BBB-related proteins than neonates. We conclude that the reactive expressional changes seen here suggest that VEGF and receptors could have a role in the excitotoxic mechanism of PNV and that such role would be less efficient in neonate rats.


Assuntos
Hipocampo/efeitos dos fármacos , Venenos de Aranha/toxicidade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Hipocampo/metabolismo , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Wistar , Aranhas , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
15.
Toxicon ; 66: 37-46, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23419593

RESUMO

Two astrocyte markers, the glial water channel aquaporin-4 (AQP4) and the glial fibrillary acidic protein (GFAP), have been implicated in several physiological and pathological conditions in the central nervous system (CNS) as well as in blood-brain barrier breakdown (BBBb). By color segmentation the immunoreactivity of both proteins, we demonstrate that the expression of AQP4 and GFAP was increased in the cerebellum of neonate (14-day-old, P14) and adult (8-week-old) rats administered Phoneutria nigriventer spider venom (PNV) known to cause perivascular edema, BBBb and convulsion. In the cerebellum's gray matter, PNV produced a major response, especially in the granular layer. Parallel increases in AQP4 and GFAP expression occurred 24 h after envenomation in the white matter of P14 and in the molecular layer of adults, as well as in the granular layer 2 h after envenomation. In the Purkinje layer there was a tendency of increased AQP4, for both, neonates (5 h), and adults (2 and 24 h). Moreover, PNV also provoked nonparallel upregulation of both markers with prevalence of upregulation of AQP 4 for P14 rats, and GFAP for adults. The major expression of both proteins was in the gray matter. The data indicates a venom effect in water/electrolyte balance in the cerebellum and the participation of AQP4 in these effects. Age-related and time-related regional differences probably reflect specificity in AQP4 distribution in different astrocytic membrane domains as well as its participation in K(+) buffering and neural activity. This study is the first to associate astrocytic AQP4 expression and reactive gliosis in a model of BBB permeability promoted by P. nigriventer venom. Our data provide compelling evidence that AQP4 expression was increased in the cerebellum of rats administered PNV.


Assuntos
Aquaporina 4/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Venenos de Aranha/toxicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade da Membrana Celular/fisiologia , Cerebelo/metabolismo , Edema/induzido quimicamente , Edema/metabolismo , Edema/patologia , Processamento de Imagem Assistida por Computador , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA