Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(17)2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39272864

RESUMEN

Diffuse large B cell lymphoma (DLBCL) is the most diagnosed, aggressive non-Hodgkin lymphoma, with ~40% of patients experiencing refractory or relapsed disease. Given the low response rates to current therapy, alternative treatment strategies are necessary to improve patient outcomes. Here, we sought to develop an easily accessible new xenograft mouse model that better recapitulates the human disease for preclinical studies. We generated two Luciferase (Luc)-EGFP-expressing human DLBCL cell lines representing the different DLBCL cell-of-origin subtypes. After intravenous injection of these cells into humanized NSG mice, we monitored the tumor growth and evaluated the organ-specific engraftment/progression period. Our results showed that human IL6-expressing NSG (NSG-IL6) mice were highly permissive for DLBCL cell growth. In NSG-IL6 mice, systemic engraftments of both U2932 activated B cell-like- and VAL germinal B cell-like-DLBCL (engraftment rate; 75% and 82%, respectively) were detected within 2nd-week post-injection. In the organ-specific ex vivo evaluation, both U2932-Luc and VAL-Luc cells were initially engrafted and expanded in the spleen, liver, and lung and subsequently in the skeleton, ovary, and brain. Consistent with the dual BCL2/MYC translocation association with poor patient outcomes, VAL cells showed heightened proliferation in human IL6-conditioned media and caused rapid tumor expansion and early death in the engrafted mice. We concluded that the U2932 and VAL cell-derived human IL6-expressing mouse models reproduced the clinical features of an aggressive DLBCL with a highly consistent pattern of tumor development. Based on these findings, NSG mice expressing human IL6 have the potential to serve as a new tool to develop DLBCL xenograft models to overcome the limitations of standard subcutaneous DLBCL xenografts.

2.
Mol Cancer Ther ; 23(3): 316-329, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-37816504

RESUMEN

Expression of the serine/threonine kinase never in mitosis gene A (NIMA)-related kinase 2 (NEK2) is essential for entry into mitosis via its role in facilitating centrosome separation. Its overactivity can lead to tumorigenesis and drug resistance through the activation of several oncogenic pathways, including AKT. Although the cancer-enabling activities of NEK2 are documented in many malignancies, including correlations with poor survival in myeloma, breast, and non-small cell lung cancer, little is known about the role of NEK2 in lymphoma. Here, in tumors from patients with diffuse large B-cell lymphoma (DLBCL), the most common, aggressive non-Hodgkin lymphoma, we found a high abundance of NEK2 mRNA and protein associated with an inferior overall survival. Using our recently developed NEK2 inhibitor, NBI-961, we discovered that DLBCL cell lines and patient-derived cells exhibit a dependency on NEK2 for their viability. This compromised cell fitness was directly attributable to efficient NEK2 inhibition and proteasomal degradation by NBI-961. In a subset of particularly sensitive DLBCL cells, NBI-961 induced G2/mitosis arrest and apoptosis. In contrast, an existing indirect NEK2 inhibitor, INH154, did not prevent NEK2 autophosphorylation, induce NEK2 proteasomal degradation, or affect cell viability. Global proteomics and phospho-proteomics revealed that NEK2 orchestrates cell-cycle and apoptotic pathways through regulation of both known and new signaling molecules. We show the loss of NEK2-sensitized DLBCL to the chemotherapy agents, doxorubicin and vincristine, and effectively suppressed tumor growth in mice. These studies establish the oncogenic activity of NEK2 in DLBCL and set the foundation for development of anti-NEK2 therapeutic strategies in this frequently refractory and relapse-prone cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Linfoma de Células B Grandes Difuso , Linfoma , Humanos , Animales , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas Relacionadas con NIMA/genética , Línea Celular Tumoral , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética
3.
Cancers (Basel) ; 15(9)2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37173940

RESUMEN

Multiple myeloma (MM) is the second most common hematological malignancy. It is a clonal B-cell disorder characterized by the proliferation of malignant plasma cells in the bone marrow, the presence of monoclonal serum immunoglobulin, and osteolytic lesions. An increasing amount of evidence shows that the interactions of MM cells and the bone microenvironment play a significant role, suggesting that these interactions may be good targets for therapy. The osteopontin-derived collagen-binding motif-bearing peptide NIPEP-OSS stimulates biomineralization and enhances bone remodeling dynamics. Due to its unique targeted osteogenic activity with a broad safety margin, we evaluated the potential of NIPEP-OSS for anti-myeloma activity using MM bone disease (MMBD) animal models. In a 5TGM1-engrafted NSG model, the survival rates of the control and treated groups were significantly different (p = 0.0014), with median survival times of 45 and 57 days, respectively. The bioluminescence analyses showed that myeloma slowly developed in the treated mice compared to the control mice in both models. NIPEP-OSS enhanced bone formation by increasing biomineralization in the bone. We also tested NIPEP-OSS in a well-established 5TGM1-engrafted C57BL/KaLwRij model. Similar to the previous model, the median survival times of the control and treated groups were significantly different (p = 0.0057), with 46 and 63 days, respectively. In comparison with the control, an increase in p1NP was found in the treated mice. We concluded that NIPEP-OSS delays mouse myeloma progression via bone formation in MMBD mouse models.

4.
Cancers (Basel) ; 13(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34503090

RESUMEN

Multiple myeloma (MM) is a plasma cell malignancy that causes an accumulation of terminally differentiated monoclonal plasma cells in the bone marrow, accompanied by multiple myeloma bone disease (MMBD). MM animal models have been developed and enable to interrogate the mechanism of MM tumorigenesis. However, these models demonstrate little or no evidence of MMBD. We try to establish the MMBD model with severe bone lesions and easily accessible MM progression. 1 × 106 luciferase-expressing 5TGM1 cells were injected into 8-12 week-old NOD SCID gamma mouse (NSG) and C57BL/KaLwRij mouse via the tail vein. Myeloma progression was assessed weekly via in vivo bioluminescence (BL) imaging using IVIS-200. The spine and femur/tibia were extracted and scanned by the micro-computer tomography for bone histo-morphometric analyses at the postmortem. The median survivals were 56 days in NSG while 44.5 days in C57BL/KaLwRij agreed with the BL imaging results. Histomorphic and DEXA analyses demonstrated that NSG mice have severe bone resorption that occurred at the lumbar spine but no significance at the femur compared to C57BL/KaLwRij mice. Based on these, we conclude that the systemic 5TGM1 injected NSG mouse slowly progresses myeloma and develops more severe MMBD than the C57BL/KaLwRij model.

5.
Front Genet ; 12: 640954, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163520

RESUMEN

Multiple myeloma (MM) is a clonal B-cell disorder characterized by the proliferation of malignant plasma cells (PCs) in the bone marrow, the presence of monoclonal serum immunoglobulin, and osteolytic lesions. It is the second most common hematological malignancy and considered an incurable disease despite significant treatment improvements. MM bone disease (MMBD) is defined as the presence of one or more osteolytic bone lesions or diffused osteoporosis with compression fracture attributable to the underlying clonal PC disorder. MMBD causes severe morbidity and increases mortality. Cumulative evidence shows that the interaction of MM cells and bone microenvironment plays a significant role in MM progression, suggesting that these interactions may be good targets for therapy. MM animal models have been developed and studied in various aspects of MM tumorigenesis. In particular, MMBD has been studied in various models, and each model has unique features. As the general features of MM animal models have been reviewed elsewhere, the current review will focus on the features of MMBD animal models.

6.
Arch Med Res ; 49(4): 226-234, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-30314650

RESUMEN

AIM OF THE STUDY: Rutin and Silibinin are active flavonoid compounds, well-known for possessing multiple biological activities. We have studied how Rutin and Silibinin in combination modulate wide range intracellular signaling cascades as evidenced by in-vitro research. Data obtained from preclinical studies provide evidence to be supportive to bridge basic and translational studies. METHODS: In this study, cytotoxic effect of Rutin and Silibinin individually and in combination on the viability of colon cancer cell line (HT-29) was revealed using the MTT assay. Mechanism involved in the cytotoxic effect were then investigated in terms of apoptosis using comet assay, DNA fragmentation and fluorescent microscopy analyses. The apoptosis associated proteins viz; Caspase-3, 8, 9, Bax, Bcl-2, p53, inflammation associated proteins viz; NFκB, IKK-α IKK-ß and MAPK pathway associated proteins viz; p38 and MK-2 were determined by western-blot and Real Time-PCR analysis. RESULTS: Results suggest that Rutin and Silibinin produce anticancer effects via induction of apoptosis as well as regulating the expressions of genes related to apoptosis, inflammation and MAPK pathway proteins more effectively in combination than individually. CONCLUSION: Our study supports the viability of developing Rutin and Silibinin in combination as a novel therapeutic prodrug for colon cancer treatment and may have a promising role in the development of new anticancer drugs in the future.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Rutina/farmacología , Silibina/farmacología , Caspasa 3 , Línea Celular Tumoral , Sinergismo Farmacológico , Células HT29 , Humanos , Proteínas Proto-Oncogénicas c-bcl-2 , Silimarina
7.
Food Chem Toxicol ; 115: 244-259, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29545143

RESUMEN

Plant phytoconstituents have been a valuable source of clinically important anticancer agents. Antioxidant and anticancerous activity of plant Curculigo orchioides Gaertn were explored In vitro antioxidant activity, antioxidant enzyme activity of oxidatively stressed tissue, and cell culture studies on human cancer cell lines HepG2, HeLa and MCF-7 were carried out. Active plant fractions were subjected to GC-MS analysis and compounds selected on the basis of their abundance were screened in silico with the help of Auto Dock 4.2 tools with pre-selected antioxidant enzymes. Curculigo orchioides Gaertn plant fractions exhibited significant antioxidant activities by virtue of scavenging of free radicals having IC50 value of ethylacetate fraction (EA) for DPPH radical scavenging assay to be 52.93 ±â€¯0.66 µg/ml. Further, antioxidant enzyme defense of mammalian tissue when treated with plant fractions revealed that enzyme concentrations were refurbished which were increased during oxidative stress. MTT assay on cell lines HepG2, HeLa and MCF-7 presented IC50 values of ethylacetate (EA) fraction as 171.23 ±â€¯2.1 µg/ml, 144.80 ±â€¯1.08 µg/ml and 153.51 µg/ml and aqueous ethylacetate (AEA) fraction as 133.44 ±â€¯1.1 µg/ml, 136.50 ±â€¯0.8 µg/ml and 145.09 µg/ml respectively. Further EA and AEA plant fractions down regulated the levels of antiapoptotic Bcl-2 expression and upregulated the expression of apoptotic proteins caspase-3 and caspase-8 through an intrinsic ROS-mediated mitochondrial dysfunction pathway. KEY MESSAGE: Key findings explained that fractions of Curculigo orchioides Gaertn inhibited oxidative stress by increasing the antioxidant enzyme content and have anticancerous potential on cancer cell lines HepG2, HeLa and MCF-7.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antioxidantes/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Curculigo/química , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Simulación por Computador , Cromatografía de Gases y Espectrometría de Masas , Células HeLa , Células Hep G2 , Humanos , Peróxido de Hidrógeno/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Células MCF-7 , Óxido Nítrico/metabolismo
8.
Arch Biochem Biophys ; 635: 8-16, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051068

RESUMEN

Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease characterized by auto-antibodies against native deoxyribonucleic acid after modification and is one of the reasons for the development of SLE. Here, we have evaluated the structural perturbations in human placental DNA by peroxynitrite using spectroscopy, thermal denaturation and high-performance liquid chromatography (HPLC). Peroxynitrite is a powerful potent bi-functional oxidative/nitrative agent that is produced both endogenously and exogenously. In experimental animals, the peroxynitrite-modified DNA was found to be highly immunogenic. The induced antibodies showed cross-reactions with different types of DNA and nitrogen bases that were modified with peroxynitrite by inhibition ELISA. The antibody activity was inhibited by approximately 89% with its immunogen as the inhibitor. The antigen-antibodies interaction between induced antibodies with peroxynitrite-modified DNA showed retarded mobility as compared to the native form. Furthermore, significantly increased binding was also observed in SLE autoantibodies with peroxynitrite-modified DNA than native form. Moreover, DNA isolated from lymphocyte of SLE patients revealed significant recognition of anti-peroxynitrite-modified DNA immunoglobulin G (IgG). Our data indicates that DNA modified with peroxynitrite presents unique antigenic determinants that may induce autoantibody response in SLE.


Asunto(s)
Autoanticuerpos/química , Autoanticuerpos/genética , Autoantígenos/química , ADN/química , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ácido Peroxinitroso/química , Autoanticuerpos/inmunología , Autoantígenos/genética , Autoantígenos/inmunología , ADN/genética , ADN/inmunología , Daño del ADN , Femenino , Humanos , Placenta/química , Embarazo , Unión Proteica
9.
Biomed Pharmacother ; 94: 265-279, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28763750

RESUMEN

Plant-derived substances (phytochemicals) are well recognized as sources of pharmacologically potent drugs in the treatment of several oxidative stress related disorders. Our study aims to evaluate the antioxidant and apoptotic effects of Glycyrrhiza glabra L. in both cell free and cell culture system. Plant fractions have been prepared with hexane, chloroform, ethyl acetate, methanol and water and their antioxidant properties are reviewed. Potent antioxidant activity has been well established in both in vitro and in silico studies which is believed to be responsible for the anticancerous nature of the plant. Results obtained indicate that methanol fraction of G. glabra L. exhibited maximum scavenging activity against DPPH and nitric oxide free radicals comparable to standard antioxidant L-AA. Administration of methanol fraction also considerably reduced the malondialdehyde produced due to lipid peroxidation in mammalian liver tissues. Moreover, the levels of antioxidant enzymes SOD, CAT, GST, GPx and GR in the oxidative stress induced tissues were refurbished significantly after treatment with plant's methanol fraction. Moreover, methanol fraction was found to be nontoxic to normal human cell line whereas it inhibited cancer cells HeLa and HepG2 considerably. Apoptosis was established by DAPI fluorescent staining and western blot analysis of pro apoptotic protein caspase-8, caspase-3 and anti-apoptotic protein Bcl-2.There is an up regulation in the levels of pro apoptotic caspase-8 and caspase-3 and down regulation of anti-apoptotic Bcl-2. Furthermore, GC-MS analysis of the methanol fraction revealed the presence of many compounds. In silico experiments using Autodock 4.2 tools showed strong affinity of plant compounds towards antioxidant enzymes (proteins) thus validating with the conclusions of antioxidant enzyme assays and establishing a role in cancer pathogenesis.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Glycyrrhiza/química , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Antioxidantes/aislamiento & purificación , Compuestos de Bifenilo/química , Supervivencia Celular/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Peroxidación de Lípido/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/enzimología , Picratos/química , Extractos Vegetales/aislamiento & purificación , Rizoma/química
10.
Int J Biol Macromol ; 97: 115-122, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28082219

RESUMEN

Aim of the studies to synthesized chitosan nanoparticles by an ionic interaction procedure. The nanoparticles were characterized by physicochemical methods like, DLS, TEM, Surface potential measurements, FT-IR and DSC. The average particle size of chitosan and carboplatin nanoparticles was found to be 277.25±11.37nm and 289.30±8.15nm and zeta potential was found to be 31±3.14mV and 33±2.15mV respectively with low polydispersity index. The maximum entrapment of carboplatin in nanoparticles was a spherical shape with a positive charge. The maximum encapsulation and loading efficiencies of carboplatin (5mg/ml) were obtained to be 58.43% and 13.27% respectively. The nanocarboplatin was better blood compatibility as compared to chitosan nanoparticles. Finally, the cytotoxic effects of the carboplatin loaded chitosan nanoparticles were tested in-vitro against breast cancer (MCF-7) cell lines. Our studies showed that the chitosan nanoparticles could be used as a promising candidate for drug delivery for the therapeutic treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carboplatino/química , Carboplatino/farmacología , Quitosano/química , Portadores de Fármacos/química , Nanopartículas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Transporte Biológico , Neoplasias de la Mama/patología , Quitosano/metabolismo , Quitosano/toxicidad , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Estabilidad de Medicamentos , Hemólisis/efectos de los fármacos , Humanos , Espacio Intracelular/metabolismo , Células MCF-7
11.
Eur J Med Chem ; 126: 705-753, 2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-27951484

RESUMEN

Benzimidazole, a fused heterocycle bearing benzene and imidazole has gained considerable attention in the field of contemporary medicinal chemistry. The moiety is of substantial importance because of its wide array of pharmacological activities. This nitrogen containing heterocycle is a part of a number of therapeutically used agents. Moreover, a number of patents concerning this moiety in the last few years further highlight its worth. The present review covers the recent work published by scientists across the globe during last five years.


Asunto(s)
Bencimidazoles/síntesis química , Química Farmacéutica/métodos , Bencimidazoles/uso terapéutico , Descubrimiento de Drogas , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/uso terapéutico
12.
Int J Biol Macromol ; 93(Pt A): 242-253, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27565296

RESUMEN

In the present study the chitosan nanoparticles (CsNPs) and curcumin loaded chitosan nanoparticles (CLCsNPs) were synthesized by tripolyphosphate (TPP) cross-linking method. The nanoparticles were prepared within a zone of appropriate chitosan and TPP concentrations. The average size of CsNPs and CLCsNPs were approximately 189±11.8nm and 197±16.8nm, exhibited a zeta potential of +76±5.6mV and +71±6.4mV respectively and drug entrapment efficiency was ≈85%. The CLCsNPs and CsNPs were further characterized by different physicochemical methods like transmission electron microscopy (TEM), dynamic light scattering (DLS), HPLC, MALDI-TOF, FT-IR, XRD and UV-vis Spectroscopy. In vitro studies revealed a fast release of ≈35% at pH 5 and ≈25% at pH 7.4 of the drug during the first 3h, followed by controlled release of curcumin over a period of 120h and sustained anti-proliferative activity of the drug in a dose and time dependent manner of CLCsNPs and combination with methyl jasmonate. The higher cytotoxicity effect of CLCsNPs may be due to their higher cellular uptake as compared to curcumin. Chitosan nanoparticles were not only stable but also a nontoxic. Our data suggested that curcumin loaded nanoformulations, therefore, might be promising candidates in cancer therapy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Quitosano/química , Curcumina/química , Curcumina/farmacología , Nanopartículas/química , Neoplasias del Cuello Uterino/patología , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Transporte Biológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quitosano/toxicidad , Curcumina/metabolismo , Curcumina/farmacocinética , Portadores de Fármacos/química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Estabilidad de Medicamentos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
Carbohydr Polym ; 151: 546-556, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27474599

RESUMEN

Chondroitin-4-sulfate (CS), a glycosaminoglycan, was used to prepare CS-capped super-paramagnetic iron oxide nanoparticles, which were further employed for loading a water-soluble chemotherapeutic agent (doxorubicin hydrochloride, DOX). CS-capped SPIONs have potential biomedical application in cancer targeting. The optimized formulation had a hydrodynamic size of 91.2±0.8nm (PDI; 0.228±0.004) and zeta potential of -49.1±1.66mV. DOX was loaded onto the formulation up to 2% (w/w) by physical interaction with CS. TEM showed nano-sized particles having a core-shell structure. XRD confirmed crystal phase of iron oxide. FT-IR conceived the interaction of iron oxide with CS as bidentate chelation and also confirmed DOX loading. Vibration sample magnetometry confirmed super-paramagnetic nature of nanoparticles, with saturation magnetization of 0.238emug(-1). In vitro release profile at pH 7.4 showed that 96.67% of DOX was released within 24h (first order kinetics). MTT assay in MCF7 cells showed significantly higher (p<0.0001) cytotoxicity for DOX in SPIONs than DOX solution (IC50 values 6.294±0.4169 and 11.316±0.1102µgmL(-1), respectively).


Asunto(s)
Antineoplásicos , Sulfatos de Condroitina , Doxorrubicina , Portadores de Fármacos , Nanopartículas de Magnetita , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Sulfatos de Condroitina/administración & dosificación , Sulfatos de Condroitina/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Células MCF-7 , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestructura , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X
14.
Toxicol Sci ; 134(2): 355-65, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23697686

RESUMEN

Paraquat (PQ), a quaternary nitrogen herbicide, is commonly used as a pesticide despite of its high toxicity. Our study evaluated the effect of subchronic PQ exposure on the neuropathology, genotoxicity, and antioxidant activity on the nervous tissue of Drosophila melanogaster. We also explored the behavioral effect of PQ on D. melanogaster. Furthermore, we attempted to validate the mechanism by evaluating PQ-induced cytotoxicity on the D-Mel2 cell lines. The fruit fly D. melanogaster serves as a feasible model to understand the mechanism of neurodegenerative diseases. Our study shows a dose-dependent PQ-induced neuropathology in the brain tissue of D. melanogaster as evidenced by hematoxylin and eosin staining, silver nitrate staining, and transmission electron microscopy. Electron microscopic study of D. melanogaster brain tissue exhibited vacuolar degeneration and significant neuronal damage across the nervous tissue structure in comparison with control. Our findings also indicate a dose-dependent locomotor impairment and decreased superoxide dismutase (SOD) specific activity in PQ-treated D. melanogaster. These PQ-induced neuroanatomical changes and decreased SOD specific activity showed a significant association with oxidative DNA damage as observed by alkaline comet assay. Additionally, we show, for the first time, a dose-dependent PQ-induced cytotoxicity in the D-Mel2 cells suggesting loss of neuronal cell viability via cytotoxic damage. Our data suggest that PQ exposure results in neurodegeneration in D. melanogaster and that fruit fly is a suitable in vivo model for correlating the neuroanatomical changes with neurotoxic damages to nervous system.


Asunto(s)
Daño del ADN , ADN/efectos de los fármacos , Herbicidas/toxicidad , Sistema Nervioso/efectos de los fármacos , Estrés Oxidativo , Paraquat/toxicidad , Animales , Ensayo Cometa , Drosophila melanogaster , Modelos Animales , Sistema Nervioso/ultraestructura , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA