Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Mol Neurobiol ; 44(1): 31, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557942

RESUMO

Glioblastoma multiforme (GBM) is the most predominant and malignant primary brain tumor in adults. Thymic stromal lymphopoietin (TSLP), a cytokine primarily generated by activated epithelial cells, has recently garnered attention in cancer research. This study was aimed to elucidate the significance of TSLP in GBM cells and its interplay with the immune system, particularly focused on granulocyte neutrophils. Our results demonstrate that the tumor produces TSLP when stimulated with epidermal growth factor (EGF) in both the U251 cell line and the GBM biopsy (GBM-b). The relevance of the TSLP function was evaluated using a 3D spheroid model. Spheroids exhibited increased diameter, volume, and proliferation. In addition, TSLP promoted the generation of satellites surrounding the main spheroids and inhibited apoptosis in U251 treated with temozolomide (TMZ). Additionally, the co-culture of polymorphonuclear (PMN) cells from healthy donors with the U251 cell line in the presence of TSLP showed a reduction in apoptosis and an increase in IL-8 production. TSLP directly inhibited apoptosis in PMN from GBM patients (PMN-p). Interestingly, the vascular endothelial growth factor (VEGF) production was elevated in PMN-p compared with PMN from healthy donors. Under these conditions, TSLP also increased VEGF production, in PMN from healthy donors. Moreover, TSLP upregulated programed death-ligand 1 (PDL-1) expression in PMN cultured with U251. On the other hand, according to our results, the analysis of RNA-seq datasets from Illumina HiSeq 2000 sequencing platform performed with TIMER2.0 webserver demonstrated that the combination of TSLP with neutrophils decreases the survival of the patient. In conclusion, our results position TSLP as a possible new growth factor in GBM and indicate its modulation of the tumor microenvironment, particularly through its interaction with PMN.


Assuntos
Glioblastoma , Linfopoietina do Estroma do Timo , Adulto , Humanos , Células Cultivadas , Citocinas/metabolismo , Neutrófilos/metabolismo , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular
2.
Nitric Oxide ; 138-139: 10-16, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37279819

RESUMO

INTRODUCTION: Gliomas represent the most prevalent form of brain tumors, among which glioblastomas are the most malignant subtype. Despite advances in comprehending their biology and treatment strategies, median survival remains disappointingly low. Inflammatory processes involving nitric oxide (NO), critically contribute to glioma formation. The inducible isoform of NO synthase (iNOS) is highly overexpressed in gliomas and has been linked to resistance against temozolomide (TMZ) treatment, neoplastic transformation, and modulation of immune response. While both in vitro and in vivo studies showed the potential of iNOS inhibitors as effective treatments for gliomas, no clinical trials on gliomas have been published. This review aims to summarize the available evidence regarding iNOS as a target for glioma treatment, focusing on clinically relevant data. METHODS: Following PRISMA guidelines, we conducted a systematic review by searching PubMed/Medline, and Embase databases in May 2023. We included studies that investigated the impact of NOS inhibitors on glioma cells using L-NMMA, CM544, PBN, 1400W or l-NAME either alone or combined with TMZ. We extracted data on the NOS inhibitor used, subtype, study setting, animal model or cell lines employed, obtained results, and safety profile. Our inclusion criteria encompassed original articles in English or Spanish, studies with an untreated control group, and a primary outcome focused on the biological effects on glioma cells. RESULTS: Out of 871 articles screened from the aforementioned databases, 37 reports were assessed for eligibility. After excluding studies that did not utilize glioma cells or address the designated outcome, 11 original articles satisfied the inclusion and exclusion criteria. Although no NOS inhibitor has been tested in a published clinical trial, three inhibitors have been evaluated using in vivo models of intracranial gliomas. l-NAME, 1400W, and CM544 were tested in vitro. Co-administration of l-NAME, or CM544 with TMZ showed superior results in vitro compared to individual agent testing. CONCLUSION: Glioblastomas remain a challenging therapeutic target. iNOS inhibitors exhibit substantial potential as treatment options for oncologic lesions, and they have demonstrated a safe toxicity profile in humans for other pathological conditions. Research endeavors should be focused on investigating their potential effects on brain tumors.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Animais , Humanos , Glioblastoma/tratamento farmacológico , NG-Nitroarginina Metil Éster/uso terapêutico , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Óxido Nítrico Sintase , Óxido Nítrico/uso terapêutico
3.
Invest New Drugs ; 40(1): 30-41, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34478029

RESUMO

Breast cancer is the leading cause of cancer death among women worldwide. For this reason, the development of new therapies is still essential. In this work we have analyzed the antitumor potential of levoglucosenone, a chiral building block derived from the pyrolysis of cellulose-containing materials such as soybean hulls, and three structurally related analogues. Employing human and murine mammary cancer models, we have evaluated the effect of our compounds on cell viability through MTS assay, apoptosis induction by acridine orange/ethidium bromide staining and/or flow cytometry and the loss of mitochondrial potential by tetramethylrhodamine methyl ester staining. Autophagy and senescence induction were also evaluated by Western blot and ß-galactosidase activity respectively. Secreted metalloproteases activity was determined by quantitative zymography. Migratory capacity was assessed by wound healing assays while invasive potential was analyzed using Matrigel-coated transwell chambers. In vivo studies were also performed to evaluate subcutaneous tumor growth and experimental lung colonization. All compounds impaired in vitro proliferation with IC50 values in a range of low micromolar. Apoptosis was identified as the main mechanism responsible for the reduction of monolayer cell content induced by the compounds without detecting modulations of autophagy or senescence processes. Two of the four compounds (levoglucosenone and its brominated variant) were able to modulate in vitro events associated with tumor progression, such as migratory potential, invasiveness, and proteases secretion. Furthermore, tumor volume and metastatic spread were significantly reduced in vivo after the treatment these two compounds. Here, we could obtain from soybean hulls, a material with almost no commercial value, a variety of chemical compounds useful for breast cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Glucose/análogos & derivados , Animais , Antineoplásicos Fitogênicos/química , Apoptose/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Celulose/química , Relação Dose-Resposta a Droga , Glucose/química , Glucose/farmacologia , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Carga Tumoral/efeitos dos fármacos
6.
Cytokine ; 84: 47-55, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27236354

RESUMO

Progress in comparative oncology promises advances in clinical cancer treatments for both companion animals and humans. In this context, feline mammary carcinoma (FMC) cells have been proposed as a suitable model to study human breast cancer. Based on our previous data about the advantages of using type I interferon gene therapy over the respective recombinant DNA derived protein, the present work explored the effects of feline interferon-ω gene (fIFNω) transfer on FMC cells. Three different cell variants derived from a single spontaneous highly aggressive FMC tumor were successfully established and characterized. Lipofection of the fIFNω gene displayed a significant cytotoxic effect on the three cell variants. The extent of the response was proportional to ROS generation, mitochondrial membrane potential disruption and calcium uptake. Moreover, a lower sensitivity to the treatment correlated with a higher malignant phenotype. Our results suggest that fIFNω lipofection could offer an alternative approach in veterinary oncology with equal or superior outcome and with less adverse effects than recombinant fIFNω therapy.


Assuntos
Interferon Tipo I/metabolismo , Neoplasias Mamárias Animais/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Cálcio/metabolismo , Gatos , Linhagem Celular Tumoral , Feminino
7.
Transl Oncol ; 13(11): 100842, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32781368

RESUMO

Most cancer cells exacerbate the pentose phosphate pathway (PPP) to enhance biosynthetic precursors and antioxidant defenses. Metformin, which is used as a first-line oral drug for the treatment of type 2 diabetes, has been proposed to inhibit the malignant progression of different types of cancers. However, metformin has shown poor efficacy as single agent in several clinical trials. Thus, the aim of the present work was to investigate whether the pharmacological inhibition of G6PDH, the first and rate-limiting enzyme of the PPP, by 6-amino nicotinamide (6-AN) potentiates the antitumoral activity of metformin on different human melanoma cell lines. Our results showed that 6-AN has sensitizing properties to metformin cytotoxicity. The combination of metformin and 6-AN decreased glucose consumption and lactate production, altered the mitochondrial potential and redox balance, and thereby blocked melanoma cell progression, directing cells to apoptosis and necrosis. To our knowledge, this is the first study describing the effect of this combination. Future preclinical studies should be performed to reveal the biological relevance of this finding.

8.
3 Biotech ; 8(8): 341, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30073126

RESUMO

Feline interferon beta is a cytokine that belongs to the type I IFN family, with antitumor, antiviral and immunomodulatory functions. In this work, recombinant feline interferon beta (rFeIFNß) was expressed in insect larvae that constitute important agronomic plagues. rFeIFNß accumulated in the hemolymph of Spodoptera frugiperda larvae infected with recombinant baculovirus and was purified by Blue-Sepharose chromatography directly from larval homogenates on day 4 post-infection. rFeIFNß was recovered after purification with a specific activity of 1 × 106 IU mg-1. By this method, we obtained 8.9 × 104 IU of purified rFeIFNß per larva. The product was biologically active in vitro, with an antiviral activity of 9.5 × 104 IU mL-1, as well as a potent antitumor activity comparable to that of the commercial FeIFNω. The glycosylation of rFeIFNß was confirmed by peptide-N-glycosidase F digestion. Our findings provide a cost-effective platform for large-scale rFeIFNß production in laboratory research or veterinary medicine applications.

9.
Oncoscience ; 4(11-12): 199-214, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29344558

RESUMO

We originated and characterized melanoma cell lines derived from tumors of two feline and two canine veterinary patients. These lines reestablished the morphology, physiology and cell heterogeneity of their respective parental tumors. We evaluated the cytotoxicity of bleomycin (BLM) alone, or combined with interferon-ß (IFN-ß) or HSVtk/GCV suicide gene (SG) lipofection on these cells. Although the four animals presented stage III disease (WHO system), SG treated feline tumors displayed stable disease in vivo, while the canine ones exhibited partial response. Their derived cell lines reflected this behavior. Feline were significantly more sensitive than canine cells to IFN-ß gene transfer. BLM improved the antitumor effects of both genes. The higher levels of reactive oxygen species (ROS) significantly correlated with membrane and DNA damages, emphasizing ROS intervention in apoptotic and necrotic cell death. After 3 days of BLM alone or combined with gene treatments, the colony forming capacity of two canine and one feline treatments survivor cells almost disappeared. Taken together, these results suggest that the treatments eradicated tumor initiating cells and support the clinical potential of the tested combinations.

10.
Res Vet Sci ; 114: 461-468, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28802138

RESUMO

Feline mammary carcinoma (FMC) is a highly aggressive pathology that has been proposed as an interesting model of breast cancer disease, especially for the hormone refractory subgroup. Recently, cancer cell metabolism has been described as a hallmark of cancer cells. Here, we investigate the effects and mechanism of metabolic modulation by metformin (MET, anti-diabetic drug), 2-deoxyglucose (2DG, hexokinase inhibitor) or a combination of both drugs, MET/2DG on two established FMC cells lines: AlRB (HER2 (3+) and Ki67<5%) and AlRATN (HER2 (-) and Ki67>15%). We found that treatments significantly decreased both FMC cells viability by up to 80%. AlRB resulted more sensitive to 2DG than AlRATN (IC50: 3.15 vs 6.32mM, respectively). The combination of MET/2DG potentiated the effects of the individually added drugs on FMC cells. In addition, MET/2DG caused an increased in intracellular oxidants, autophagic vesicles and completely inhibited colony formation. Conversely, only MET significantly altered plasma membrane integrity, presented late apoptotic/necrotic cells and increased both glucose consumption and lactate concentration. Our results support further studies to investigate the potential use of this metabolic modulation approach in a clinical veterinary setting.


Assuntos
Antineoplásicos/farmacologia , Desoxiglucose/metabolismo , Metabolismo Energético/efeitos dos fármacos , Neoplasias Mamárias Animais/metabolismo , Metformina/farmacologia , Animais , Gatos , Linhagem Celular Tumoral/metabolismo , Sobrevivência Celular/efeitos dos fármacos
11.
Biomed Pharmacother ; 83: 290-301, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27399807

RESUMO

We explored the potential of a chemogene therapy combination to eradicate melanoma tumor initiating cells, key producers of recurrence and metastatic spread. Three new human melanoma cell lines, two obtained from lymph nodes and one from spleen metastasis were established and characterized. They were cultured as monolayers and spheroids and, in both spatial configurations they displayed sensitivity to single treatments with bleomycin (BLM) or human interferon-ß (hIFNß) gene or herpes simplex virus thymidine kinase/ganciclovir suicide gene (SG) lipofection. However, the combination of bleomycin with SG or hIFNß gene transfer displayed greater antitumor efficacy. The three cell lines exhibited a proliferative behavior consistent with melan A and gp100 melanoma antigens expression, and BRAF V600E mutation. BLM and both genetic treatments increased the fraction of more differentiated and treatment-sensitive cells. Simultaneously, they significantly decreased the sub-population of tumor initiating cells. There was a significant correlation between the cytotoxicity of treatments with BLM and gene transfer and the fraction of cells exhibiting (i) high proliferation index, and (ii) high intracellular levels of reactive oxygen species. Conversely, the fraction of cells surviving to our treatments closely paralleled their (i) colony and (ii) melanosphere forming capacity. A very significant finding was that the combination of BLM with SG or hIFNß gene almost abrogated the clonogenic capacity of the surviving cells. Altogether, the results presented here suggest that the combined chemo-gene treatments are able to eradicate tumor initiating cells, encouraging further studies aimed to apply this strategy in the clinic.


Assuntos
Bleomicina/uso terapêutico , Genes Transgênicos Suicidas , Terapia Genética , Interferon beta/genética , Melanoma/tratamento farmacológico , Células-Tronco Neoplásicas/patologia , Neoplasias Cutâneas/tratamento farmacológico , Biomarcadores Tumorais/metabolismo , Bleomicina/farmacologia , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Clonais , Humanos , Imuno-Histoquímica , Melanoma/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/patologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
12.
Toxicology ; 207(3): 501-10, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15664276

RESUMO

The possibility that Tl(OH)3, the main Tl3+ specie present in water solutions, could interfere with the normal functioning of the glutathione-dependent antioxidant defense system was investigated. For this purpose, we used both the purified components of this system and rat brain cytosolic fractions. Tl(OH)3 (1-25 microM) significantly decreased the content of reduced glutathione (GSH) in both experimental systems, caused by GSH oxidation. In the same range of concentrations Tl(OH)3 inhibited glutathione peroxidase (GPx) activity in both models, using cumene hydroperoxide as the substrate. No alterations in the capacity of GPx activity to metabolize H2O2 were observed. Both in purified GR as well as in the cytosolic fraction, Tl(OH)3 (1-5 microM) inhibited GR activity, with a partial recovery of the activity at higher concentrations. While Tl(OH)3 inhibited the GR diaphorase activity of purified GR, in a concentration (1-25 microM) dependent manner, this effect was only observed in the cytosolic fractions at the highest concentration assessed (25 microM). Results indicate that, similarly to previous findings for Tl+ and Tl3+, Tl(OH)3 also alters the glutathione-dependent antioxidant defense system. The observed alterations of this important antioxidant protective pathway by the major Tl3+ specie in water solutions could be one mechanism involved in the oxidative stress associated to Tl-intoxication.


Assuntos
Citosol/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Glutationa/metabolismo , Tálio/toxicidade , Animais , Encéfalo , Química Encefálica , Catalase/metabolismo , Fracionamento Celular , Citosol/enzimologia , Relação Dose-Resposta a Droga , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , NADP/metabolismo , Oxirredução , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
13.
Biomed Pharmacother ; 72: 44-51, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26054674

RESUMO

A local gene therapy scheme for the delivery of type I interferons could be an alternative for the treatment of melanoma. We evaluated the cytotoxic effects of interferon-ß (IFNß) gene lipofection on tumor cell lines derived from three human cutaneous and four canine mucosal melanomas. The cytotoxicity of human IFNß gene lipofection resulted higher or equivalent to that of the corresponding addition of the recombinant protein (rhIFNß) to human cells. IFNß gene lipofection was not cytotoxic for only one canine melanoma cell line. When cultured as monolayers, three human and three canine IFNß-lipofected melanoma cell lines displayed a remarkable bystander effect. As spheroids, the same six cell lines were sensitive to IFNß gene transfer, two displaying a significant multicell resistance phenotype. The effects of conditioned IFNß-lipofected canine melanoma cell culture media suggested the release of at least one soluble thermolabile cytotoxic factor that could not be detected in human melanoma cells. By using a secretion signal-free truncated human IFNß, we showed that its intracellular expression was enough to induce cytotoxicity in two human melanoma cell lines. The lower cytoplasmatic levels of reactive oxygen species detected after intracellular IFNß expression could be related to the resistance displayed by one human melanoma cell line. As IFNß gene transfer was effective against most of the assayed melanomas in a way not limited by relatively low lipofection efficiencies, the clinical potential of this approach is strongly supported.


Assuntos
Técnicas de Transferência de Genes , Interferon beta/genética , Melanoma/genética , Animais , Efeito Espectador , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Cães , Humanos , Espaço Intracelular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/genética , Temperatura , Transfecção , Transgenes , beta-Galactosidase/genética
14.
Hum Gene Ther ; 26(6): 367-76, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25762364

RESUMO

We present here a nonviral immunogene therapy trial for canine malignant melanoma, an aggressive disease displaying significant clinical and histopathological overlapping with human melanoma. As a surgery adjuvant approach, it comprised the co-injection of lipoplexes bearing herpes simplex virus thymidine kinase and canine interferon-ß genes at the time of surgery, combined with the periodic administration of a subcutaneous genetic vaccine composed of tumor extracts and lipoplexes carrying the genes of human interleukin-2 and human granulocyte-macrophage colony-stimulating factor. Following complete surgery (CS), the combined treatment (CT) significantly raised the portion of local disease-free canine patients from 11% to 83% and distant metastases-free (M0) from 44% to 89%, as compared with surgery-only-treated controls (ST). Even after partial surgery (PS), CT better controlled the systemic disease (M0: 82%) than ST (M0: 48%). Moreover, compared with ST, CT caused a significant 7-fold (CS) and 4-fold (PS) rise of overall survival, and >17-fold (CS) and >13-fold (PS) rise of metastasis-free survival. The dramatic increase of PS metastasis-free survival (>1321 days) and CS recurrence- and metastasis-free survival (both >2251 days) demonstrated that CT was shifting a rapidly lethal disease into a chronic one. In conclusion, this surgery adjuvant CT was able of significantly delaying or preventing postsurgical recurrence and distant metastasis, increasing disease-free and overall survival, and maintaining the quality of life. The high number of canine patients involved in CT (301) and the extensive follow-up (>6 years) with minimal or absent toxicity warrant the long-term safety and efficacy of this treatment. This successful clinical outcome justifies attempting a similar scheme for human melanoma.


Assuntos
Vacinas Anticâncer/farmacologia , Doenças do Cão/terapia , Terapia Genética/métodos , Interferon beta/genética , Melanoma/veterinária , Adjuvantes Imunológicos/farmacologia , Animais , Terapia Combinada , Citocinas/metabolismo , Doenças do Cão/mortalidade , Doenças do Cão/cirurgia , Cães , Feminino , Genes Transgênicos Suicidas , Masculino , Melanoma/mortalidade , Melanoma/cirurgia , Melanoma/terapia , Resultado do Tratamento
15.
Free Radic Res ; 38(9): 977-84, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15621716

RESUMO

We investigated the hypothesis that thallium (Tl) interactions with the glutathione-dependent antioxidant defence system could contribute to the oxidative stress associated with Tl toxicity. Working in vitro with reduced glutathione (GSH), glutathione reductase (GR) or glutathione peroxidase (GPx) in solution, we studied the effects of Tl+ and Tl3+ (1-25 microM) on: (a) the amount of free GSH, investigating whether the metal binds to GSH and/or oxidizes it; (b) the activity of the enzyme GR, that catalyzes GSH regeneration; and (c) the enzyme GPx, that reduces hydroperoxide at expense of GSH oxidation. We found that, while Tl+ had no effect on GSH concentration, Tl3+ oxidized it. Both cations inhibited the reduction of GSSG by GR and the diaphorase activity of this enzyme. In addition, Tl3+ per se oxidized NADPH, the cofactor of GR. The effects of Tl on GPx activity depended on the metal charge: Tl+ inhibited GPx when cumene hydroperoxide (CuOOH) was the substrate, while Tl(3+)-mediated GPx inhibition occurred with both substrates. The present results show that Tl interacts with all the components of GSH/GSSG antioxidant defence system. Alterations of this protective pathway could be partially responsible for the oxidative stress associated with Tl toxicity.


Assuntos
Antioxidantes/química , Dissulfeto de Glutationa/química , Glutationa Peroxidase/química , Glutationa Redutase/química , Glutationa/química , Tálio/química , Antioxidantes/análise , Derivados de Benzeno/análise , Derivados de Benzeno/química , Cátions/química , Glutationa/análise , Dissulfeto de Glutationa/análise , Glutationa Peroxidase/análise , Glutationa Redutase/análise , Peróxido de Hidrogênio/análise , Peróxido de Hidrogênio/química , NADP/análise , NADP/química , Oxirredução
16.
Chem Phys Lipids ; 122(1-2): 159-63, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12598047

RESUMO

We investigated whether Al(3+)-mediated changes in membrane fluidity can affect the activity of prokaryotic enzymes phospholipase C (PLC) and phospholipase C-phosphatidyl inositol specific (PI-PLC) in liposomes of phosphatidyl choline (PC), PC:phosphatidyl inositol (PI), or PC and polyphosphoinositides (PPI). Al(3+) (10-100 microM) promoted membrane rigidification, evaluated with the probes 1,6-diphenyl-1,3,5-hexatriene and Laurdan, and followed the order: PC:PPI>PC:PI>PC. Al(3+) (25 and 50 microM) did not affect PLC-mediated hydrolysis of PC, PI and PIP(2), but stimulated PIP hydrolysis (48.6%). PI-PLC did not affect PC, PI, and PIP concentrations, but caused a 67% decrease in PIP(2). Al(3+) significantly inhibited PIP(2) hydrolysis in a concentration-dependent (25-50 microM) manner. Results suggest that the inhibition of PIP(2) hydrolysis by Al(3+) could be partially due to a higher lipid packing induced by Al(3+) which could affect the interaction between the enzyme and its substrate.


Assuntos
Alumínio/farmacologia , Fluidez de Membrana/efeitos dos fármacos , Fosfatidilinositol Diacilglicerol-Liase/metabolismo , Fosfolipases Tipo C/metabolismo , Polarização de Fluorescência , Hidrólise , Lipossomos , Fosfoinositídeo Fosfolipase C
17.
Int Immunopharmacol ; 22(1): 167-75, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24973616

RESUMO

In an aggressive B16-F10 murine melanoma model, we evaluated the effectiveness and antitumor mechanisms triggered by a surgery adjuvant treatment that combined a local suicide gene therapy (SG) with a subcutaneous genetic vaccine (Vx) composed of B16-F10 cell extracts and lipoplexes carrying the genes of human interleukin-2 and murine granulocyte and macrophage colony stimulating factor. Pre-surgical SG treatment, neither alone nor combined with Vx was able to slow down the fast evolution of this tumor. After surgery, both SG and SG + Vx treatments, significantly prevented (in 50% of mice) or delayed (in the remaining 50%) post-surgical recurrence, as well as significantly prolonged recurrence-free (SG and SG + Vx) and overall median survival (SG + Vx). The treatment induced the generation of a pseudocapsule wrapping and separating the tumor from surrounding host tissue. Both, SG and the subcutaneous Vx, induced this envelope that was absent in the control group. On the other hand, PET scan imaging of the SG + Vx group suggested the development of an effective systemic immunostimulation that enhanced (18)FDG accrual in the thymus, spleen and vertebral column. When combined with surgery, direct intralesional injection of suicide gene plus distal subcutaneous genetic vaccine displayed efficacy and systemic antitumor immune response without host toxicity. This suggests the potential value of the assayed approach for clinical purposes.


Assuntos
Vacinas Anticâncer , Procedimentos Cirúrgicos Dermatológicos , Terapia Genética/métodos , Imunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutâneas/terapia , Pele/patologia , Animais , Extratos Celulares , Genes Transgênicos Suicidas/genética , Fator Estimulador de Colônias de Granulócitos/genética , Humanos , Interleucina-2/genética , Fator Estimulador de Colônias de Macrófagos/genética , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Recidiva Local de Neoplasia , Transplante de Neoplasias , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia
18.
Biomed Pharmacother ; 67(4): 269-75, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23453489

RESUMO

Bleomycin is a chemotherapeutic agent barely diffusible through the plasmatic membrane. We evaluated DNA/cationic lipids complexes (lipoplexes) as mediators of its uptake in four spontaneous canine melanoma derived cell lines (Ak, Bk, Br and Rkb). Cell survival after lipofection plus or minus bleomycin was determined by the acid phosphatase method and the cellular uptake of lipoplexes, carrying the E. coli ß-galactosidase gene, was evidenced by SYBR Green I staining. The four cell lines resulted sensitive to the bleomycin/lipoplexes system in both spatial configurations. Survival rates values were lower than 20% in monolayers of the four tested lines and lower than 30% in three lines (Ak, Bk and Rkb) when grown as spheroids. The sensitization to bleomycin depended on lipoplexes in Ak and Rkb while Bk (in both spatial configurations) and Br (as monolayers) were sensitive to bleomycin alone. Although some degree of sensitivity to bleomycin was induced by cationic lipids alone in Ak and Rkb monolayers, the maximal bleomycin effects appeared in the presence of lipoplexes. The sensitization was independent of transcriptional activity. The co-administration of lipoplexes diminished bleomycin IC50: 10-fold in Ak and Rkb monolayers; and sensitized the Ak and Rkb resistant spheroids. The bleomycin cytotoxic effects depended on lipoplexes concentration and diminished when cells were incubated at 8°C. Our results suggest that lipoplexes sensitize cells to bleomycin, increasing its uptake by an active transport mechanism, such as endocytosis. The bleomycin/lipoplexes system appears as a promising combination of chemotherapy and non-viral cancer gene therapy.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Bleomicina/administração & dosagem , DNA/administração & dosagem , Melanoma/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacologia , Transporte Biológico Ativo , Bleomicina/farmacocinética , Bleomicina/farmacologia , Cátions , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doenças do Cão/tratamento farmacológico , Doenças do Cão/patologia , Cães , Resistencia a Medicamentos Antineoplásicos , Endocitose , Terapia Genética/métodos , Concentração Inibidora 50 , Lipídeos/química , Lipossomos , Melanoma/patologia , Melanoma/veterinária , Temperatura
19.
Res Vet Sci ; 91(2): 230-4, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21300385

RESUMO

Eleven soft tissue- and five osteosarcoma canine patients were subjected to: (i) periodic subcutaneous injection of irradiated xenogeneic cells secreting hGM-CSF and hIL-2 mixed with allogeneic or autologous tumor homogenates; and (ii) injections of cIFN-ß and HSVtk-carrying lipoplexes and ganciclovir, marginal (after surgery) and/or intratumoral (in the case of partial tumor resection, local relapse or small surface tumors). This treatment alone (4 patients) or as surgery adjuvant (12 patients), was safe and well tolerated. In those patients presenting local disease (6/11), the suicide gene plus cIFN-ß treatment induced local antitumor activity evidenced by the objective responses (3 complete, 2 partial) and stable diseases (2). In addition, the treatment prevented or delayed local relapse, regional metastases (lymph nodes developed only in 3/16) and distant metastases (0/16), suggesting a strong systemic antitumor immunity. The most encouraging result was the long survival times of 10 patients (>1 year, with good quality of life).


Assuntos
Apoptose/genética , Vacinas Anticâncer/uso terapêutico , Citocinas/genética , Doenças do Cão/terapia , Terapia Genética/veterinária , Interferon beta/genética , Sarcoma/veterinária , Animais , Terapia Combinada/veterinária , Doenças do Cão/cirurgia , Cães , Feminino , Ganciclovir/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interleucina-2/genética , Masculino , Sarcoma/cirurgia , Sarcoma/terapia , Simplexvirus/enzimologia , Timidina Quinase/genética , Resultado do Tratamento
20.
Arch Biochem Biophys ; 417(2): 235-43, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12941306

RESUMO

The hypothesis that thallium (Tl) interaction with membrane phospholipids could result in the alteration of membrane physical properties was investigated. Working with liposomes composed of brain phosphatidylcholine and phosphatidylserine, we found that Tl(+), Tl(3+), and Tl(OH)(3) (0.5-25 microM): (a) increased membrane surface potential, (b) decreased the fluidity of the anionic regions of the membrane, in association with an increased fluidity in the cationic regions, and (c) promoted the rearrangement of lipids through lateral phase separation. The magnitude of these effects followed the order Tl(3+), Tl(OH)(3)>Tl(+). In addition, Tl(3+) also decreased the hydration of phospholipid polar headgroups and induced membrane permeabilization. The present results show that Tl interacts with membranes inducing major alterations in the rheology of the bilayer, which could be partially responsible for the neurotoxic effects of this metal.


Assuntos
Lipossomos/química , Fosfatidilcolinas/química , Fosfatidilserinas/química , Tálio/química , Química Encefálica , Lipossomos/síntese química , Substâncias Macromoleculares , Fluidez de Membrana , Fusão de Membrana , Potenciais da Membrana , Permeabilidade , Eletricidade Estática , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA