Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 9918, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37336969

ABSTRACT

Obesity in adolescents has reached epidemic proportions and is associated with the inflammatory response and viral infections. The aim of this study was to understand the profile of inflammatory cytokines and chemokines associated with the inflammatory response and metabolic syndrome (MetS) in obese adolescents with positive serology for adenovirus 36 (ADV36). Thirty-six overweight, 36 obese, and 25 severe obesity adolescents aged 10 to 16 years were included in the study. The following variables were analyzed: sex, age, body mass index (BMI), blood pressure, total cholesterol and fractions, triglycerides, glucose, serum cytokine concentrations, and ADV36 antibodies. Cytokines and chemokines were quantified by cytometry and ADV36 serology was determined by enzyme-linked immunosorbent assay (ELISA). The results showed higher levels of the cytokines interleukin-1beta (IL-1ß), IL-6, IL-10 and of the chemokine interferon-gamma-inducible protein 10 (IP-10) in severe obesity adolescents compared to the obese and overweight groups, as well as in the group with MetS compared to the group without this syndrome. The frequency of ADV36-positive individuals did not differ between groups. The findings revealed differences in BMI between the obese and severe obesity groups versus the overweight group in the presence of positivity for ADV36, suggesting an association with weight gain and possibly MetS installation.


Subject(s)
Adenoviridae Infections , Metabolic Syndrome , Obesity, Morbid , Pediatric Obesity , Adolescent , Humans , Adenoviridae , Overweight , Cytokines , Adenoviridae Infections/epidemiology , Body Mass Index
2.
Mater Sci Eng C Mater Biol Appl ; 116: 111254, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32806261

ABSTRACT

Multiple-wall carbon nanotubes (CNTs) were functionalized with polyethyleneimine in order to incorporate paclitaxel (PTX), the first line chemotherapeutic agent for prostate cancer. These particles were then covered with antibodies for the prostate-specific membrane antigen (PSMA), to address them to prostate cancer cells. LNCaP prostate cancer cells (PSMA+), HCT-116 and CaCo-2 colon cancer cells (PSMA-), as well as human peripheral monocytes and lymphocytes (PSMA-), were in vitro exposed to fluorescent CNT composites. The interaction/adherence of those composites to target cells was analyzed by fluorescence microscopy and flow cytometry, showing a diffuse interaction of CNTs and CNT-PTX with all cell types. Analysis of cytotoxicity revealed that both prostate (PSMA+) and colorectal cancer cells (PSMA-) were more susceptible to PTX complexed with CNTs than to pure PTX or CNTs alone, while the incorporation of anti-PSMA (CNT-PTX-PSMA) improved the toxicity on LNCaP cells but not on PSMA- targets. No toxicity was observed in human monocytes and lymphocytes but composites induced phenotypical changes in monocytes. Our results demonstrate the feasibility of using anti-PSMA antibody to address drug-loaded CNT to cancer cells as a strategy for improving the effectiveness of antineoplastic agents.


Subject(s)
Nanotubes, Carbon , Prostatic Neoplasms , Antibodies, Monoclonal , Caco-2 Cells , Cell Line, Tumor , Humans , Male , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy
3.
Int Immunopharmacol ; 84: 106495, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32298965

ABSTRACT

Autophagy is an important mechanism for tumor escape, allowing tumor cells to recover from the damage induced by chemotherapy, radiation therapy, and immunotherapy and contributing to the development of resistance. The pharmacological inhibition of autophagy contributes to increase the efficacy of antineoplastic agents. Exposing tumor cells to low concentrations of select autophagy-inducing antineoplastic agents increases their immunogenicity and enhances their ability to stimulate dendritic cell (DC) maturation. We tested whether the application of an autophagy-inhibiting agent, chloroquine (CQ), in combination with low concentrations of 5-fluorouracil (5-FU) increases the ability of tumor cells to induce DC maturation. DCs sensitized with the lysate of HCT-116 cells previously exposed to such a combination enhanced the DC maturation/activation ability. These matured DCs also increased the allogeneic responsiveness of both CD4+ and CD8+ T cells, which showed a greater proliferative response than those from DCs sensitized with control lysates. The T cells expanded in such cocultures were CD69+ and PD-1- and produced higher levels of IFN-γ and lower levels of IL-10, consistent with the preferential activation of Th1 cells. Cocultures of autologous DCs and lymphocytes improved the generation of cytotoxic T lymphocytes, as assessed by the expression of CD107a, perforin, and granzyme B. The drug combination increased the expression of genes related to the CEACAM family (BECN1, ATGs, MAPLC3B, ULK1, SQSTM1) and tumor suppressors (PCBP1). Furthermore, the decreased expression of genes related to metastasis and tumor progression (BNIP3, BNIP3L, FOSL2, HES1, LAMB3, LOXL2, NDRG1, P4HA1, PIK3R2) was noted. The combination of 5-FU and CQ increases the ability of tumor cells to drive DC maturation and enhances the ability of DCs to stimulate T cell responses.


Subject(s)
Autophagy/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chloroquine/pharmacology , Dendritic Cells/metabolism , Antimetabolites, Antineoplastic/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Fluorouracil/pharmacology , HCT116 Cells , Humans , Lymphocyte Activation/drug effects , Th1 Cells/drug effects , Th1 Cells/metabolism , Transcriptional Activation/drug effects
4.
Cancer J ; 25(3): 165-177, 2019.
Article in English | MEDLINE | ID: mdl-31135523

ABSTRACT

The clear cell subtype of kidney cancer encompasses most renal cell carcinoma cases and is associated with the loss of von Hippel-Lindau gene function or expression. Subsequent loss or mutation of the other allele influences cellular stress responses involving nutrient and hypoxia sensing. Autophagy is an important regulatory process promoting the disposal of unnecessary or degraded cellular components, tightly linked to almost all cellular processes. Organelles and proteins that become damaged or that are no longer needed in the cell are sequestered and digested in autophagosomes upon fusing with lysosomes, or alternatively, released via vesicular exocytosis. Tumor development tends to disrupt the regulation of the balance between this process and apoptosis, permitting prolonged cell survival and increased replication. Completed trials of autophagic inhibitors using hydroxychloroquine in combination with other anticancer agents including rapalogues and high-dose interleukin 2 have now been reported. The complex nature of autophagy and the unique biology of clear cell renal cell carcinoma warrant further understanding to better develop the next generation of relevant anticancer agents.


Subject(s)
Autophagy , Carcinoma, Renal Cell/metabolism , Endothelium/metabolism , Kidney Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Autophagy/genetics , Biomarkers , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/pathology , Clinical Trials as Topic , Cytoskeleton/metabolism , Disease Susceptibility , Drug Discovery , Drug Screening Assays, Antitumor , Endothelium/drug effects , Endothelium/pathology , High-Throughput Nucleotide Sequencing , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/etiology , Kidney Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mitophagy/drug effects , Mitophagy/genetics , Molecular Imaging , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
Med Mycol ; 56(7): 911-915, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29228322

ABSTRACT

Microorganisms killing by dendritic cells (DCs) is an important effector mechanism during innate immune response, as it can avoid dissemination of infection during migration of these cells toward draining lymph nodes. However, this function depends on pattern recognition receptors (PRRs) to which the microorganism will bind in these cells. Regarding this, TLR9 activation, by stimulating the oxidative metabolism, induces increase in microbicidal activity of these cells. Accordingly, we showed that DCs treatment with a TLR9 agonist results in an increase in fungicidal activity of these cells against the fungus Paracoccidioides brasiliensis (Pb), which however, was not associated to higher H2O2 levels.


Subject(s)
Dendritic Cells/immunology , Microbial Viability , Paracoccidioides/immunology , Toll-Like Receptor 9/metabolism , Cells, Cultured , Healthy Volunteers , Humans , Hydrogen Peroxide/metabolism
6.
Cancer Biother Radiopharm ; 32(8): 302-308, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29053415

ABSTRACT

AIM: Considering the central role of dendritic cells (DCs) on the development of an antitumor immune response, in this study we used a murine model to evaluate how DC transfection with drug-treated tumor cell RNA changes their phenotype, and whether transfection enhances the in vivo effectiveness of a DC-based antitumor vaccine. MATERIALS AND METHODS: MC-38 colorectal tumor cells were pretreated with the minimum effective concentration of 5-fluorouracil (5-FU), then their total RNA was extracted and transfected into DCs. These DCs were inoculated into C57Bl/6 mice bearing subcutaneous MC-38 tumor. RESULTS: DC transfection with drug-treated tumor RNA increases the percentages of CD40+ (from 37.6% to 61.4%), CD86+ (from 39.8% to 53.4%), and major histocompatibility complex class II+ (from 51.2% to 75.3%) cells, whereas significantly increases the in vivo generation of interferon-γ producer lymphocytes. CONCLUSION: These results reinforce our view that treatment of tumor cells with 5-FU induces transcriptional changes that can be transferred to DCs by RNA transfection, enhancing their ability to stimulate an antitumor response.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Dendritic Cells/drug effects , Fluorouracil/administration & dosage , RNA, Neoplasm/genetics , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic/drug effects , Transfection/methods , Treatment Outcome , Xenograft Model Antitumor Assays
7.
J Pharm Pharmacol ; 69(11): 1625-1633, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28776672

ABSTRACT

OBJECTIVES: The effects of propolis and phenolic compounds (caffeic acid - Caf; dihydrocinnamic acid - Cin; p-coumaric acid - Cou) in the same quantity found in our propolis sample were investigated on human laryngeal epidermoid carcinoma (HEp-2) cells. METHODS: Cell viability, apoptosis/necrosis and cell cycle arrest, P53 and CASPASE-3 gene expression, generation of reactive oxygen species (ROS) and the ability of propolis to induce doxorubicin (DOX) efflux using a P-glycoprotein (P-gp) inhibitor (verapamil) were assayed. KEY FINDINGS: Propolis exerted a cytotoxic effect on HEp-2 cells, whereas isolated compounds had no effect on cell viability. Higher concentrations were tested and Caf induced late apoptosis or necrosis in HEp-2 cells, while propolis induced apoptosis, both probably due to ROS generation. P53 expression was downregulated by propolis but not by Caf. CASPASE-3 expression was correlated with induction of both early and late apoptosis, with both propolis and Caf alone upregulating its expression. Propolis induced cell cycle arrest at G2/M phase and Caf at S phase. Propolis but not Caf may act as a P-gp inhibitor by modulating P-gp activity and inhibiting DOX efflux. CONCLUSIONS: Propolis exerted cytotoxic effects on HEp-2 cells, and the mechanisms are discussed, showing its potential as an antitumour drug.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Caffeic Acids/pharmacology , Laryngeal Neoplasms/drug therapy , Propolis/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Apoptosis/drug effects , Brazil , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Laryngeal Neoplasms/pathology , Phenols/pharmacology , Reactive Oxygen Species/metabolism , Verapamil/pharmacology , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...