Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Extracell Vesicles ; 11(12): e12294, 2022 12.
Article in English | MEDLINE | ID: mdl-36537293

ABSTRACT

Transforming growth factor ß (TGFß) is a major component of tumor-derived small extracellular vesicles (TEX) in cancer patients. Mechanisms utilized by TGFß+ TEX to promote tumor growth and pro-tumor activities in the tumor microenvironment (TME) are largely unknown. TEX produced by head and neck squamous cell carcinoma (HNSCC) cell lines carried TGFß and angiogenesis-promoting proteins. TGFß+ TEX stimulated macrophage chemotaxis without a notable M1/M2 phenotype shift and reprogrammed primary human macrophages to a pro-angiogenic phenotype characterized by the upregulation of pro-angiogenic factors and functions. In a murine basement membrane extract plug model, TGFß+ TEX promoted macrophage infiltration and vascularization (p < 0.001), which was blocked by using the TGFß ligand trap mRER (p < 0.001). TGFß+ TEX injected into mice undergoing the 4-nitroquinoline-1-oxide (4-NQO)-driven oral carcinogenesis promoted tumor angiogenesis (p < 0.05), infiltration of M2-like macrophages in the TME (p < 0.05) and ultimately tumor progression (p < 0.05). Inhibition of TGFß signaling in TEX with mRER ameliorated these pro-tumor activities. Silencing of TGFß emerges as a critical step in suppressing pro-angiogenic functions of TEX in HNSCC.


Subject(s)
Extracellular Vesicles , Head and Neck Neoplasms , Humans , Animals , Mice , Squamous Cell Carcinoma of Head and Neck , Transforming Growth Factor beta/metabolism , Extracellular Vesicles/metabolism , Macrophages/metabolism , Neovascularization, Pathologic/genetics , Phenotype , Tumor Microenvironment
2.
Arch Physiol Biochem ; 128(1): 223-230, 2022 Feb.
Article in English | MEDLINE | ID: mdl-31595805

ABSTRACT

The aim of this study was to investigate the ability of tannic acid (TA) in preventing memory deficits and neurochemical alterations observed in a model for Sporadic Dementia of Alzheimer's Type. Rats were treated with TA (30 mg/kg) daily for 21 days, and subsequently received intracerebroventricular injection of streptozotocin (STZ). We observed that STZ induced learning and memory impairments; however, treatment with TA was able to prevent these effects. In cerebral cortex and hippocampus, STZ induced an increase in acetylcholinesterase activity, reduced Na+, K+-ATPase activity and induced oxidative stress increasing thiobarbituric acid-reactive substances, nitrites and reactive oxygen species levels and reducing the activity of antioxidant enzymes. Treatment with TA was able in prevent the major of these neurochemical alterations. In conclusion, TA prevented memory deficits, alterations in brain enzyme activities, and oxidative damage induced by STZ. Thus, TA can be an interesting strategy in the prevention of Sporadic Alzheimer's Disease.


Subject(s)
Alzheimer Disease , Acetylcholinesterase/metabolism , Adenosine Triphosphatases , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Animals , Disease Models, Animal , Maze Learning , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar , Streptozocin/toxicity , Tannins
3.
mBio ; 12(4): e0165721, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34253061

ABSTRACT

Extracellular vesicles (EVs) have recently garnered attention for their participation in host-microbe interactions in pneumococcal infections. However, the effect of EVs on the host immune system remain poorly understood. Our studies focus on EVs produced by Streptococcus pneumoniae (pEVs), and reveal that pEVs are internalized by macrophages, T cells, and epithelial cells. In vitro, pEVs induce NF-κB activation in a dosage-dependent manner and polarize human macrophages to an alternative (M2) phenotype. In addition, pEV pretreatment conditions macrophages to increase bacteria uptake and such macrophages may act as a reservoir for pneumococcal cells by increasing survival of the phagocytosed bacteria. When administered systemically in mice, pEVs induce cytokine release; when immobilized locally, they recruit lymphocytes and macrophages. Taken together, pEVs emerge as critical contributors to inflammatory responses and tissue damage in mammalian hosts. IMPORTANCE Over the last decade, pathogen-derived extracellular vesicles (EVs) have emerged as important players in several human diseases. Therefore, a thorough understanding of EV-mediated mechanisms could provide novel insights into vaccine/therapeutic development. A critical question in the field is: do pathogen-derived EVs help the pathogen evade the harsh environment in the host or do they help the host to mount a robust immune response against the pathogen? This study is a step towards answering this critical question for the Gram-positive pathogen, Streptococcus pneumoniae. Our study shows that while S. pneumoniae EVs (pEVs) induce inflammatory response both in vitro and in vivo, they may also condition the host macrophages to serve as a reservoir for the bacteria.


Subject(s)
Extracellular Vesicles/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , Streptococcus pneumoniae/immunology , Animals , Female , Macrophages/classification , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Phagocytosis , Phenotype , Pneumococcal Infections/immunology , Signal Transduction/immunology
4.
Mol Cancer Ther ; 20(6): 1102-1111, 2021 06.
Article in English | MEDLINE | ID: mdl-33850003

ABSTRACT

TGFß is a key regulator of oral squamous cell carcinoma (OSCC) progression, and its potential role as a therapeutic target has been investigated with a limited success. This study evaluates two novel TGFß inhibitors as mono or combinatorial therapy with anti-PD-L1 antibodies (α-PD-L1 Ab) in a murine OSCC model. Immunocompetent C57BL/6 mice bearing malignant oral lesions induced by 4-nitroquinoline 1-oxide (4-NQO) were treated for 4 weeks with TGFß inhibitors mRER (i.p., 50 µg/d) or mmTGFß2-7m (10 µg/d delivered by osmotic pumps) alone or in combination with α-PD-L1 Abs (7× i.p. of 100 µg/72 h). Tumor progression and body weight were monitored. Levels of bioactive TGFß in serum were quantified using a TGFß bioassay. Tissues were analyzed by immunohistology and flow cytometry. Therapy with mRER or mmTGFß2-7m reduced tumor burden (P < 0.05) and decreased body weight loss compared with controls. In inhibitor-treated mice, levels of TGFß in tumor tissue and serum were reduced (P < 0.05), whereas they increased with tumor progression in controls. Both inhibitors enhanced CD8+ T-cell infiltration into tumors and mRER reduced levels of myeloid-derived suppressor cells (P < 0.001). In combination with α-PD-L1 Abs, tumor burden was not further reduced; however, mmTGFß2-7m further reduced weight loss (P < 0.05). The collagen-rich stroma was reduced by using combinatorial TGFß/PD-L1 therapies (P < 0.05), enabling an accelerated lymphocyte infiltration into tumor tissues. The blockade of TGFß signaling by mRER or mmTGFß2-7m ameliorated in vivo progression of established murine OSCC. The inhibitors promoted antitumor immune responses, alone and in combination with α-PD-L1 Abs.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Mouth Neoplasms/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/pathology , Disease Progression , Humans , Immune Checkpoint Inhibitors/pharmacology , Longitudinal Studies , Mice , Mouth Neoplasms/pathology
5.
Int J Mol Sci ; 21(11)2020 Jun 02.
Article in English | MEDLINE | ID: mdl-32498400

ABSTRACT

Interactions between tumor cells and tumor-associated macrophages (TAMs) are critical for glioblastoma progression. The TAMs represent up to 30% of the glioblastoma mass. The role of TAMs in tumor progression and in the mechanisms underlying tumor growth remain unclear. Using an in vitro model resembling the crosstalk between macrophages and glioblastoma cells, we show that glioblastoma-derived exosomes (GBex) reprogram M1 (mediate pro-inflammatory function) and M2 (mediate anti-inflammatory function) macrophages, converting M1 into TAMs and augmenting pro-tumor functions of M2 macrophages. In turn, these GBex-reprogrammed TAMs, produce exosomes decorated by immunosuppressive and tumor-growth promoting proteins. TAM-derived exosomes disseminate these proteins in the tumor microenvironment (TME) promoting tumor cell migration and proliferation. Mechanisms underlying the promotion of glioblastoma growth involved Arginase-1+ exosomes produced by the reprogrammed TAMs. A selective Arginase-1 inhibitor, nor-NOHA reversed growth-promoting effects of Arginase-1 carried by TAM-derived exosomes. The data suggest that GBex-reprogrammed Arginase-1+ TAMs emerge as a major source of exosomes promoting tumor growth and as a potential therapeutic target in glioblastoma.


Subject(s)
Arginase/metabolism , Brain Neoplasms/physiopathology , Exosomes/metabolism , Glioblastoma/physiopathology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Immunosuppressive Agents/metabolism , Inflammation , Phenotype , Tumor Microenvironment
6.
Angiogenesis ; 23(4): 599-610, 2020 11.
Article in English | MEDLINE | ID: mdl-32419057

ABSTRACT

RATIONALE: One hallmark of tumor-derived exosomes (TEX) is the promotion of cancer progression by stimulating angiogenesis. This study was performed to evaluate the role of adenosine receptors in TEX-induced angiogenesis. METHODS: TEX produced by UMSCC47 head and neck cancer cell line were isolated by mini size exclusion chromatography (mini-SEC). Enzymatic activity of ectonucleotidases CD39/CD73 carried by TEX was measured by HPLC. Adenosine content of TEX was measured by UPLC-MS/MS. Primary human macrophages were co-incubated with TEX or exosomes derived from the plasma of head and neck cancer patients and their marker expression profile was analyzed by flow cytometry. The macrophage secretome was analyzed by angiogenesis arrays. The in vitro angiogenic potential of TEX was evaluated in endothelial growth studies. Results were validated in vivo using basement membrane extract plug assays in A1R-/-, A2AR-/- and A2BR-/- rats. Vascularization was analyzed by hemoglobin quantification and immunohistology with vessel and macrophage markers. RESULTS: TEX carried enzymatically active CD39/CD73 and adenosine. TEX promoted A2BR-mediated polarization of macrophages toward an M2-like phenotype (p < 0.05) and enhanced their secretion of angiogenic factors. Growth of endothelial cells was stimulated directly by TEX and indirectly via macrophage-reprogramming dependent on A2BR signaling (p < 0.01). In vivo, TEX stimulated the formation of defined vascular structures and macrophage infiltration. This response was absent in A2BR-/- rats (p < 0.05). CONCLUSION: This report provides the first evidence for adenosine production by TEX to promote angiogenesis via A2BR. A2BR antagonism emerges as a potential strategy to block TEX-induced angiogenesis.


Subject(s)
Exosomes/metabolism , Head and Neck Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor, Adenosine A2B/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Proliferation , Cellular Reprogramming , Exosomes/ultrastructure , Female , Head and Neck Neoplasms/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/metabolism , Male , Models, Biological , Phenotype , Rats
7.
Purinergic Signal ; 16(2): 231-240, 2020 06.
Article in English | MEDLINE | ID: mdl-32440820

ABSTRACT

Exosomes, small-sized extracellular vesicles, carry components of the purinergic pathway. The production by cells of exosomes carrying this pathway remains poorly understood. Here, we asked whether type 1, 2A, or 2B adenosine receptors (A1Rs, A2ARs, and A2BRs, respectively) expressed by producer cells are involved in regulating exosome production. Preglomerular vascular smooth muscle cells (PGVSMCs) were isolated from wildtype, A1R-/-, A2AR-/-, and A2BR-/- rats, and exosome production was quantified under normal or metabolic stress conditions. Exosome production was also measured in various cancer cells treated with pharmacologic agonists/antagonists of A1Rs, A2ARs, and A2BRs in the presence or absence of metabolic stress or cisplatin. Functional activity of exosomes was determined in Jurkat cell apoptosis assays. In PGVSMCs, A1R and A2AR constrained exosome production under normal conditions (p = 0.0297; p = 0.0409, respectively), and A1R, A2AR, and A2BR constrained exosome production under metabolic stress conditions. Exosome production from cancer cells was reduced (p = 0.0028) by the selective A2AR agonist CGS 21680. These exosomes induced higher levels of Jurkat apoptosis than exosomes from untreated cells or cells treated with A1R and A2BR agonists (p = 0.0474). The selective A2AR antagonist SCH 442416 stimulated exosome production under metabolic stress or cisplatin treatment, whereas the selective A2BR antagonist MRS 1754 reduced exosome production. Our findings indicate that A2ARs suppress exosome release in all cell types examined, whereas effects of A1Rs and A2BRs are dependent on cell type and conditions. Pharmacologic targeting of cancer with A2AR antagonists may inadvertently increase exosome production from tumor cells.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Exosomes/drug effects , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Exosomes/metabolism , Male , Phenethylamines/pharmacology , Rats , Receptor, Adenosine A1/drug effects , Receptor, Adenosine A2A/drug effects , Tumor Cells, Cultured/metabolism
8.
Pharm Dev Technol ; 25(4): 408-415, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31847658

ABSTRACT

Introduction: Glioblastoma (GB) is the most common malignant brain tumor and is characterized by high invasiveness, poor prognosis, and limited therapeutic options. Silencing gene expression, through the use of small interfering RNA (siRNA), has been proposed as an alternative to conventional cancer therapy. Here, we evaluated the potential of CD73 as a new therapeutic target, since it is overexpressed in solid tumors and has emerged as a promising target to control GB progression.Methods: A cationic nanoemulsion (NE) as an intravenous siRNA-CD73 delivery system was developed and its effect on C6 glioma cell viability was determined.Results: The nanostructured system was effective in complexing oligonucleotides for delivery to target cells. In addition, we observed that the NE-siRNA-CD73 complex was effective in reducing CD73 protein levels and AMPase activity, which were related to decreased C6 glioma cell viability.Conclusions: These findings indicate the potential of siRNA-CD73-loaded cationic NE as a therapeutic alternative for glioma treatment.


Subject(s)
5'-Nucleotidase/genetics , Glioma/therapy , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cations/chemistry , Cell Line, Tumor , Cells, Cultured , Drug Carriers/chemistry , Emulsions/chemistry , Glioma/genetics , RNA, Small Interfering/genetics , RNAi Therapeutics , Rats
9.
Metab Brain Dis ; 35(2): 283-293, 2020 02.
Article in English | MEDLINE | ID: mdl-31773434

ABSTRACT

Glioblastoma is a devastating tumor affecting the central nervous system with infiltrative capacity, high proliferation rate and chemoresistance. Therefore, it is urgent to find new therapeutic alternatives that improve this prognosis. Herein, we focused on tannic acid (TA) a polyphenol with antioxidant and antiproliferative activities. In this work, the antitumor and antioxidant effects of TA on rat (C6) glioblastoma cells and their cytotoxicity relative to primary astrocyte cultures were evaluated in vitro. Cells were exposed to TA of 6.25 to 75 µM for 24, 48 and/or 72 h. In addition, colony formation, migration and cell adhesion were analyzed and flow cytometry was used to analyze cell death and cell cycle. Next, the action of TA was evaluated in a preclinical glioblastoma model performed on Wistar rats. In this protocol, the animals were treated with a dose of 50 mg/kg/day TA for 15 days. Our results demonstrated that TA induced in vitro selective antiglioma activity, not demonstrating cytotoxicity in astrocyte culture. It induced cell death by apoptosis and cell cycle arrest, reducing formation and size of colonies, cell migration/adhesion and showing to be a potential antioxidant. Interestingly, the antiglioma effect was also observed in vivo, as TA decreased tumor volume by 55%, accompanied by an increase in the area of intratumoral necrosis and infiltration of lymphocytes without causing systemic damage. To the best of our knowledge, this is the first study to report TA activity in a GBM preclinical model. Thus, this natural compound is promising as a treatment for glioblastoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Tannins/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Female , Glioblastoma/pathology , Male , Rats , Rats, Wistar , Tannins/pharmacology
10.
Curr Protoc Immunol ; 127(1): e91, 2019 12.
Article in English | MEDLINE | ID: mdl-31763776

ABSTRACT

A method for isolation of exosomes from tumor cell supernatants or cancer patients' plasma is presented. Tumor-derived exosomes (TEX) are defined as a subset of extracellular vesicles (EVs) sized at 30 to 150 nm and originating from multivesicular bodies (MVBs). The method utilizes size exclusion chromatography (SEC) for recovery of exosomes from cell-line supernatants or cancer patients' plasma. The recovered exosomes are morphologically intact, aggregate-free, and functionally competent. Their molecular content parallels that of the parent tumor cells and they carry various immunoregulatory ligands known to modulate functions of immune cells. All exosomes isolated from tumor cell lines are TEX, while those isolated from plasma of cancer patients have to be fractionated into TEX and non-TEX. Mini-SEC allows for exosome isolation and recovery in quantities sufficient for molecular profiling, functional studies, and, in the case of plasma, further fractionation into TEX and non-TEX. The mini-SEC method can also be used for comparative studies of the exosome content in serial specimens of cancer patients' body fluids. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Exosomes/chemistry , Neoplasms/chemistry , Animals , Cell Fractionation , Cell Line, Tumor , Chromatography, Gel , Exosomes/immunology , Humans , Mice , Neoplasms/immunology , Neoplasms/pathology
11.
Cancer Lett ; 467: 85-95, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31593802

ABSTRACT

The role of CD44 in progression of head and neck squamous cell carcinoma (HNSCC) has been controversial. The goal of this study was to study the effects of CD44(+) tumor cells on the initial stages of tumor angiogenesis and to evaluate CD44 as a potential marker of tumor angiogenesis. The CD44 gene expression was studied using the Cancer Genome Atlas (TCGA) Head and Neck Cancer data base. Expression levels of CD44 and of microvascular density (MVD) markers were assessed by immunohistochemistry performed with tissue microarrays in a cohort of 49 HNSCC patients, 11 patients with dysplasia and 12 control oral mucosa tissues. The 4-nitroquinoline-1-oxide oral carcinogenesis mouse model was used to study CD44 expression during carcinogenesis. Gelatin sponges seeded with CD44(+), CD44(-) and unsorted cancer cells suspended in Matrigel were implanted in NOD/SCID mice into a dorsal skinfold chamber and compared to non-seeded sponges as controls. Angiogenic response was assessed by intravital microscopy. In the TCGA analysis, CD44 gene expression correlated with various pro-angiogenic genes. In human HNSCC tissues, CD44 expression was upregulated and was associated with blood vessels, although no correlation between MVD and CD44 expression was found. During oral carcinogenesis CD44 expression was upregulated. In dorsal skinfold chambers, CD44(+) cells showed a significantly higher MVD than CD44(-) or unsorted cells (p < 0.001). The results indicate that CD44(+) cells contain pro-angiogenic factors and stimulate tumor angiogenesis in HNSCC. Thus, CD44 might emerge as a potential angiogenic biomarker and a therapeutic target for anti-angiogenic therapies.


Subject(s)
Head and Neck Neoplasms/blood supply , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Squamous Cell Carcinoma of Head and Neck/blood supply , Up-Regulation , 4-Nitroquinoline-1-oxide/adverse effects , Animals , Case-Control Studies , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Tissue Array Analysis
12.
Biomed Chromatogr ; 33(10): e4615, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31166608

ABSTRACT

A high-performance liquid chromatography method for temozolomide (TMZ) determination in complex biological matrices was developed and validated for application in in vitro, ex vivo and in vivo studies of new nanotechnology-based systems for TMZ nasal delivery. The method was able to quantify TMZ in nanoemulsions, following cellular uptake, in the porcine nasal mucosa and in mouse plasma and brain. Analyses were performed on a C18 column at 35°C, under UV detection at 330 nm. The mobile phase was methanol-acetic acid 0.5% (30:70, v/v), eluted at an isocratic flow rate of 1.1 mL/min. The method was found to be specific, precise, accurate, robust and linear (0.05 to 5 µg/mL) for TMZ determination in all matrices. No interference of TMZ degradation products was found under various stress conditions such as acidic, alkaline, oxidative, light and thermal exposure, demonstrating stability. The method was applied for the quantification of TMZ in different matrices, i.e. the efficiency of nanoemulsions in vitro in increasing TMZ cellular uptake, ex vivo TMZ permeation and retention in the porcine nasal mucosa tissue, and for in vivo TMZ quantification in mouse brain following intranasal nanoemulsion administration compared with free TMZ.


Subject(s)
Chromatography, High Pressure Liquid/methods , Temozolomide , Administration, Intranasal , Animals , Cell Line, Tumor , Drug Stability , Emulsions/administration & dosage , Emulsions/chemistry , Emulsions/pharmacokinetics , Limit of Detection , Linear Models , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Reproducibility of Results , Spectrophotometry, Ultraviolet , Swine , Temozolomide/administration & dosage , Temozolomide/analysis , Temozolomide/chemistry , Temozolomide/pharmacokinetics
13.
J Enzyme Inhib Med Chem ; 34(1): 197-203, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30482059

ABSTRACT

A series of nineteen benzothiazin-4-ones from N-(3-aminopropyl) piperidine, 4-(2-aminoethyl)morpholine or 1-(2-aminoethyl)piperidine, aliphatic or aromatic aldehyde and thiosalicylic acid, were synthesized in good yields by multicomponent one-pot reactions. The solvent was toluene and this efficient procedure afforded the desired heterocycles in 5 h. Identification and characterization were achieved by NMR and GC-MS techniques. In vitro AChE activities of all compounds were evaluated in cerebral cortex and hippocampus of rats and in general, the results in cortex were more promising than hippocampus. The benzothiazinone 5Bd showed the best AChE inhibition activity IC50 8.48 µM (cortex) and IC50 39.80 µM (hippocampus). The cytotoxicity of seven compounds in MCR-5 human fibroblast cell by SRB test in 24 h were evaluated and 5Bd suggest preliminary safety, showing no cytotoxicity at 100 µM. Finally, these important findings could be a starting point for the development of new AChE inhibitors agents and will provide the basis for new studies.


Subject(s)
Acetylcholinesterase/metabolism , Benzothiadiazines/pharmacology , Cholinesterase Inhibitors/pharmacology , Animals , Benzothiadiazines/chemical synthesis , Benzothiadiazines/chemistry , Cells, Cultured , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Drug Design , Fibroblasts/drug effects , Humans , Male , Molecular Structure , Rats , Rats, Wistar , Structure-Activity Relationship
14.
Pharm Dev Technol ; 23(4): 400-406, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28521578

ABSTRACT

The present study showed the development of nanocapsules containing the association of the coenzyme Q10 and vitamin E acetate and the evaluation of their effect on in vitro cells culture of malignant glioma and melanoma. In order to investigate if nanocapsules are able to protect coenzyme Q10 from degradation under UVC radiation, a photostability study was carried out. For this, three concentrations of vitamin E acetate were evaluated (1%, 2%, or 3%). Nanocapsules presented suitable physicochemical characteristics and were able to protect coenzyme Q10 from photodegradation. In addition, this protection was influenced by higher vitamin E acetate concentrations, attributing to this oil an important role on coenzyme Q10 photostabilization. Regarding to in vitro citotoxicity assay, nanocapsules containing coenzyme Q10 and 2% vitamin E significantly reduced glioma and melanoma cell viability in 61% and 66%, respectively. In this sense, these formulations represent interesting platforms for the delivery of coenzyme Q10 and vitamin E acetate, presenting effect on the reduction of malignant cells viability.


Subject(s)
Antineoplastic Agents/administration & dosage , Nanocapsules/chemistry , Polymers/chemistry , Ubiquinone/analogs & derivatives , Vitamin E/administration & dosage , Antineoplastic Agents/pharmacology , Antioxidants/administration & dosage , Antioxidants/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Stability , Glioma/drug therapy , Humans , Melanoma/drug therapy , Photolysis , Ubiquinone/administration & dosage , Ubiquinone/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Vitamins/administration & dosage , Vitamins/pharmacology
15.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2652-2662, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28713019

ABSTRACT

BACKGROUND: Glioblastomas are the most devastating brain tumor characterized by chemoresistance development and poor prognosis. Macrophages are a component of tumor microenvironment related to glioma malignancy. The relation among inflammation, innate immunity and cancer is accepted; however, molecular and cellular mechanisms mediating this relation and chemoresistance remain unresolved. OBJECTIVE: Here we evaluated whether glioma sensitive or resistant to temozolomide (TMZ) modulate macrophage polarization and inflammatory pathways associated. The impact of glioma-macrophage crosstalk on glioma proliferation was also investigated. METHODS: GL261 glioma chemoresistance was developed by exposing cells to increasing TMZ concentrations over a period of 6months. Mouse peritoneal macrophages were exposed to glioma-conditioned medium or co-cultured directly with glioma sensitive (GL) or chemoresistant (GLTMZ). Macrophage polarization, in vitro and in vivo glioma proliferation, redox parameters, ectonucleotidase activity and ATP cytotoxicity were performed. RESULTS: GLTMZ cells were more effective than GL in induce M2-like macrophage polarization and in promote a strong immunosuppressive environment characterized by high IL-10 release and increased antioxidant potential, which may contribute to glioma chemoresistance and proliferation. Interestingly, macrophage-GLTMZ crosstalk enhanced in vitro and in vivo proliferation of chemoresistant cells, decreased ectonucleotidase activities, which was followed by increased macrophage sensitivity to ATP induced death. CONCLUSIONS: Results suggest a differential macrophage modulation by GLTMZ cells, which may favor the maintenance of immunosuppressive tumor microenvironment and glioma proliferation. GENERAL SIGNIFICANCE: The induction of immunosuppressive environment and macrophage education by chemoresistant gliomas may be important for tumor recovery after chemotherapy and could be considered to overcome chemoresistance development.


Subject(s)
Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Inflammation/drug therapy , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Polarity/drug effects , Dacarbazine/administration & dosage , Disease Models, Animal , Glioma/metabolism , Glioma/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Receptors, Purinergic/genetics , Temozolomide , Tumor Microenvironment/drug effects
16.
Chem Biol Interact ; 266: 1-9, 2017 Mar 25.
Article in English | MEDLINE | ID: mdl-28174097

ABSTRACT

Glioblastoma multiforme (GBM) is the worst form of primary brain tumor, which has a high rate of infiltration and resistance to radiation and chemotherapy, resulting in poor prognosis for patients. Recent studies show that thiazolidinones have a wide range of pharmacological properties including antimicrobial, anti-inflammatory, anti-oxidant and anti-tumor. Here, we investigate the effect antiglioma in vitro of a panel of sixteen synthetic 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones where 13 of these decreased the viability of glioma cells 30-65% (100 µM) compared with controls. The most promising compounds such as 4d, 4l, 4m and 4p promoted glioma reduction of viability greater than 50%, were further tested at lower concentrations (12.5, 25, 50 and 100 µM). Also, the data showed that the compounds 4d, 4l, 4m and 4p induced cell death primarily through necrosis and late apoptosis mechanisms. Interestingly, none of these 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones were cytotoxic for primary astrocytes, which were used as a non-transformed cell model, indicating selectivity. Our results also show that the treatment with sub-therapeutic doses of 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones (4d, 4l and 4p) reduced in vivo glioma growth as well as malignant characteristics of implanted tumors such as intratumoral hemorrhage and peripheral pseudopalisading. Importantly, 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones treatment did not induce mortality or peripheral damage to animals. Finally, 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones also changed the nitric oxide metabolism which may be associated with reduced growth and malignity characteristics of gliomas. These data indicates for the first time the therapeutic potential of synthetic 2-aryl-3-((piperidin-1-yl)ethyl)thiazolidin-4-ones to GBM treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Glioblastoma/pathology , Models, Biological , Thiazolidines/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Rats , Rats, Wistar
17.
Mol Cell Biochem ; 424(1-2): 69-78, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27752805

ABSTRACT

Methionine is an essential amino acid involved in critical metabolic process, and regulation of methionine flux through metabolism is important to supply this amino acid for cell needs. Elevation in plasma methionine commonly occurs due to mutations in methionine-metabolizing enzymes, such as methionine adenosyltransferase. Hypermethioninemic patients exhibit clinical manifestations, including neuronal and liver disorders involving inflammation and tissue injury, which pathophysiology is not completely established. Here, we hypothesize that alterations in macrophage inflammatory response may contribute to deleterious effects of hypermethioninemia. To this end, macrophage primary cultures were exposed to methionine (1 mM) and/or its metabolite methionine sulfoxide (0.5 mM), and M1/proinflammatory or M2/anti-inflammatory macrophage polarization was evaluated. In addition, inflammation-related pathways including oxidative stress parameters, as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activities; reactive oxygen species (ROS) production, and purinergic signaling, as ATP/ADP/AMPase activities, were investigated. Methionine and/or methionine sulfoxide induced M1/classical macrophage activation, which is related to proinflammatory responses characterized by increased iNOS activity and TNF-α release. Further experiments showed that treatments promoted alterations on redox state of macrophages by differentially modulated SOD and CAT activities and ROS levels. Finally, methionine and/or methionine sulfoxide treatment also altered the extracellular nucleotide metabolism, promoting an increase of ATPase/ADPase activities in macrophages. In conclusion, these findings contribute to better understand the participation of proinflammatory responses in cell injury observed in hypermethioninemic patients.


Subject(s)
Macrophages/metabolism , Methionine/analogs & derivatives , Methionine/pharmacology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Animals , Catalase/metabolism , Glutathione Peroxidase/metabolism , Male , Mice , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
18.
Colloids Surf B Biointerfaces ; 130: 272-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25935266

ABSTRACT

This study aimed to prepare pomegranate seed oil nanoemulsions containing ketoprofen using pullulan as a polymeric stabilizer, and to evaluate antitumor activity against in vitro glioma cells. Formulations were prepared by the spontaneous emulsification method and different concentrations of pullulan were tested. Nanoemulsions presented adequate droplet size, polydispersity index, zeta potential, pH, ketoprofen content and encapsulation efficiency. Nanoemulsions were able to delay the photodegradation profile of ketoprofen under UVC radiation, regardless of the concentration of pullulan. In vitro release study indicates that nanoemulsions were able to release approximately 95.0% of ketoprofen in 5h. Free ketoprofen and formulations were considered hemocompatible at 1 µg/mL, in a hemolysis study, for intravenous administration. In addition, a formulation containing the highest concentration of pullulan was tested against C6 cell line and demonstrated significant activity, and did not reduce fibroblasts viability. Thus, pullulan can be considered an interesting excipient to prepare nanostructured systems and nanoemulsion formulations can be considered promising alternatives for the treatment of glioma.


Subject(s)
Emulsions/chemistry , Glucans/chemistry , Ketoprofen/chemistry , Nanoparticles/chemistry , Plant Oils/chemistry , 3T3 Cells , Administration, Intravenous , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Stability , Glioma/metabolism , Glioma/pathology , Glioma/prevention & control , Hemolysis/drug effects , Ketoprofen/administration & dosage , Ketoprofen/pharmacokinetics , Kinetics , Lythraceae/chemistry , Mice , Microscopy, Electron, Scanning , Molecular Structure , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Photolysis/radiation effects , Rats , Seeds/chemistry , Ultraviolet Rays
19.
Invest New Drugs ; 31(6): 1424-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24072435

ABSTRACT

Glioblastoma multiforme (GBM) is the worst and most common brain tumor, characterized by high proliferation and invasion rates. Nanoparticles of biodegradable polymers for anticancer drug delivery have attracted interest in recent years since they provide targeted delivery and may overcame the obstacle imposed by blood-brain barrier. Here we investigated the antitumoral effect of ketoprofen-loaded nanocapsules (Keto-NC) treatment on in vitro and in vivo glioma progression. We observed that Keto-NC treatment decreased selectively the cell viability of a panel of glioma cell lines, while did not exhibited toxicity to astrocytes. We further demonstrate that the treatment with sub-therapeutic dose of Keto-NC reduced the in vivo glioma growth as well as reduced the malignity characteristics of implanted tumors. Keto-NC treatment improved the weight, the locomotion/exploration behavior of glioma-bearing rats. Importantly, Keto-NC treatment neither induced mortality or peripheral damage. Finally, Ketoprofen also altered the extracellular nucleotide metabolism of peripheral lymphocytes, suggesting that antiinflammatory effects of ketoprofen could also be associated with the modulation of the adenine nucleotide metabolism in lymphocytes. Data indicate at first time the potential of Keto-NC as a promising therapeutic alterative to GBM treatment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Ketoprofen/administration & dosage , Nanocapsules/administration & dosage , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Glioma/pathology , Humans , Male , Rats , Rats, Wistar , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...