Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Ups J Med Sci ; 1292024.
Article in English | MEDLINE | ID: mdl-38571882

ABSTRACT

Deeper analysis of molecular mechanisms arising in tumor cells is an unmet need to provide new diagnostic and therapeutic strategies to prevent and treat tumors. The transforming growth factor ß (TGF-ß) signaling has been steadily featured in tumor biology and linked to poor prognosis of cancer patients. One pro-tumorigenic mechanism induced by TGF-ß is the epithelial-to-mesenchymal transition (EMT), which can initiate cancer dissemination, enrich the tumor stem cell population, and increase chemoresistance. TGF-ß signals via SMAD proteins, ubiquitin ligases, and protein kinases and modulates the expression of protein-coding and non-coding RNA genes, including those encoding larger than 500 nt transcripts, defined as long non-coding RNAs (lncRNAs). Several reports have shown lncRNAs regulating malignant phenotypes by directly affecting epigenetic processes, transcription, and post-transcriptional regulation. Thus, this review aims to update and summarize the impact of TGF-ß signaling on the expression of lncRNAs and the function of such lncRNAs as regulators of TGF-ß signaling, and how these networks might impact specific hallmarks of cancer.


Subject(s)
Neoplasms , RNA, Long Noncoding , Humans , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Neoplasms/genetics , Signal Transduction , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic
2.
Pharmaceuticals (Basel) ; 16(12)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38139812

ABSTRACT

Anaplastic thyroid cancer (ATC) is an aggressive form of thyroid cancer (TC), accounting for 50% of total TC-related deaths. Although therapeutic approaches against TC have improved in recent years, the survival rate remains low, and severe adverse effects are commonly reported. However, unexplored alternatives based on natural compounds, such as lysicamine, an alkaloid found in plants with established cytotoxicity against breast and liver cancers, offer promise. Therefore, this study aimed to explore the antineoplastic effects of lysicamine in papillary TC (BCPAP) and ATC (HTH83 and KTC-2) cells. Lysicamine treatment reduced cell viability, motility, colony formation, and AKT activation while increasing the percentage of necrotic cells. The absence of caspase activity confirmed apoptosis-independent cell death. Necrostatin-1 (NEC-1)-mediated necrosome inhibition reduced lysicamine-induced necrosis in KTC-2, suggesting necroptosis induction via a reactive oxygen species (ROS)-independent mechanism. Additionally, in silico analysis predicted lysicamine target proteins, particularly those related to MAPK and TGF-ß signaling. Our study demonstrated lysicamine's potential as an antineoplastic compound in ATC cells with a proposed mechanism related to inhibiting AKT activation and inducing cell death.

3.
Redox Biol ; 64: 102773, 2023 08.
Article in English | MEDLINE | ID: mdl-37300955

ABSTRACT

PURPOSE: Glioblastoma (GBM) is the most common type of adult brain tumor with extremely poor survival. Cystathionine-gamma lyase (CTH) is one of the main Hydrogen Sulfide (H2S) producing enzymes and its expression contributes to tumorigenesis and angiogenesis but its role in glioblastoma development remains poorly understood. METHODS: and Principal Results: An established allogenic immunocompetent in vivo GBM model was used in C57BL/6J WT and CTH KO mice where the tumor volume and tumor microvessel density were blindly measured by stereological analysis. Tumor macrophage and stemness markers were measured by blinded immunohistochemistry. Mouse and human GBM cell lines were used for cell-based analyses. In human gliomas, the CTH expression was analyzed by bioinformatic analysis on different databases. In vivo, the genetic ablation of CTH in the host led to a significant reduction of the tumor volume and the protumorigenic and stemness transcription factor sex determining region Y-box 2 (SOX2). The tumor microvessel density (indicative of angiogenesis) and the expression levels of peritumoral macrophages showed no significant changes between the two genotypes. Bioinformatic analysis in human glioma tumors revealed that higher CTH expression is positively correlated to SOX2 expression and associated with worse overall survival in all grades of gliomas. Patients not responding to temozolomide have also higher CTH expression. In mouse or human GBM cells, pharmacological inhibition (PAG) or CTH knockdown (siRNA) attenuates GBM cell proliferation, migration and stem cell formation frequency. MAJOR CONCLUSIONS: Inhibition of CTH could be a new promising target against glioblastoma formation.


Subject(s)
Glioblastoma , Mice , Humans , Animals , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Mice, Inbred C57BL , Temozolomide , Cell Line , Cell Line, Tumor
4.
Sci Rep ; 12(1): 21113, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36477472

ABSTRACT

Glioblastoma (GBM) is the most aggressive and common primary malignant brain tumor with limited available therapeutic approaches. Despite improvements in therapeutic options for GBM patients, efforts to develop new successful strategies remain as major unmet medical needs. Based on the cytotoxic properties of aporphine compounds, we evaluated the biological effect of 12 compounds obtained through total synthesis of ( ±)-apomorphine hydrochloride (APO) against GBM cells. The compounds 2,2,2-trifluoro-1-(1-methylene-3,4-dihydroisoquinolin-2(1H)-yl)ethenone (A5) and ( ±)-1-(10,11-dimethoxy-6a,7-dihydro-4H-dibenzo[de,g]quinolin-6(5H)-yl)ethenone (C1) reduced the viability of GBM cells, with 50% inhibitory concentration ranging from 18 to 48 µM in patient-derived GBM cultures. Our data show that APO, A5 or C1 modulate the expression of DNA damage and apoptotic markers, impair 3D-gliomasphere growth and reduce the expression of stemness markers. Potential activity and protein targets of A5, C1 or APO were predicted in silico based on PASS and SEA software. Dopamine receptors (DRD1 and 5), CYP2B6, CYP2C9 and ABCB1, whose transcripts were differentially expressed in the GBM cells, were among the potential A5 or C1 target proteins. Docking analyses (HQSAR and 3D-QSAR) were performed to characterize possible interactions of ABCB1 and CYP2C9 with the compounds. Notably, A5 or C1 treatment, but not temozolomide (TMZ), reduced significantly the levels of extracellular ATP, suggesting ABCB1 negative regulation, which was correlated with stronger cytotoxicity induced by the combination of TMZ with A5 or C1 on GBM cells. Hence, our data reveal a potential therapeutic application of A5 and C1 as cytotoxic agents against GBM cells and predicted molecular networks that can be further exploited to characterize the pharmacological effects of these isoquinoline-containing substances.


Subject(s)
Temozolomide , Humans , Temozolomide/pharmacology
5.
Front Cell Dev Biol ; 10: 849938, 2022.
Article in English | MEDLINE | ID: mdl-35493080

ABSTRACT

Complexity in mechanisms that drive cancer development and progression is exemplified by the transforming growth factor ß (TGF-ß) signaling pathway, which suppresses early-stage hyperplasia, yet assists aggressive tumors to achieve metastasis. Of note, several molecules, including mRNAs, non-coding RNAs, and proteins known to be associated with the TGF-ß pathway have been reported as constituents in the cargo of extracellular vesicles (EVs). EVs are secreted vesicles delimited by a lipid bilayer and play critical functions in intercellular communication, including regulation of the tumor microenvironment and cancer development. Thus, this review aims at summarizing the impact of EVs on TGF-ß signaling by focusing on mechanisms by which EV cargo can influence tumorigenesis, metastatic spread, immune evasion and response to anti-cancer treatment. Moreover, we emphasize the potential of TGF-ß-related molecules present in circulating EVs as useful biomarkers of prognosis, diagnosis, and prediction of response to treatment in cancer patients.

6.
J Cell Physiol ; 237(1): 743-762, 2022 01.
Article in English | MEDLINE | ID: mdl-34350982

ABSTRACT

The role of liver kinase B1 (LKB1) in glioblastoma (GBM) development remains poorly understood. LKB1 may regulate GBM cell metabolism and has been suggested to promote glioma invasiveness. After analyzing LKB1 expression in GBM patient mRNA databases and in tumor tissue via multiparametric immunohistochemistry, we observed that LKB1 was localized and enriched in GBM tumor cells that co-expressed SOX2 and NESTIN stemness markers. Thus, LKB1-specific immunohistochemistry can potentially reveal subpopulations of stem-like cells, advancing GBM patient molecular pathology. We further analyzed the functions of LKB1 in patient-derived GBM cultures under defined serum-free conditions. Silencing of endogenous LKB1 impaired 3D-gliomasphere frequency and promoted GBM cell invasion in vitro and in the zebrafish collagenous tail after extravasation of circulating GBM cells. Moreover, loss of LKB1 function revealed mitochondrial dysfunction resulting in decreased ATP levels. Treatment with the clinically used drug metformin impaired 3D-gliomasphere formation and enhanced cytotoxicity induced by temozolomide, the primary chemotherapeutic drug against GBM. The IC50 of temozolomide in the GBM cultures was significantly decreased in the presence of metformin. This combinatorial effect was further enhanced after LKB1 silencing, which at least partially, was due to increased apoptosis. The expression of genes involved in the maintenance of tumor stemness, such as growth factors and their receptors, including members of the platelet-derived growth factor (PDGF) family, was suppressed after LKB1 silencing. The defect in gliomasphere growth caused by LKB1 silencing was bypassed after supplementing the cells with exogenous PFDGF-BB. Our data support the parallel roles of LKB1 in maintaining mitochondrial homeostasis, 3D-gliomasphere survival, and hindering migration in GBM. Thus, the natural loss of, or pharmacological interference with LKB1 function, may be associated with benefits in patient survival but could result in tumor spread.


Subject(s)
AMP-Activated Protein Kinase Kinases/metabolism , Brain Neoplasms , Glioblastoma , Metformin , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Metformin/pharmacology , Neoplastic Stem Cells/pathology , Protein Kinases/genetics , Temozolomide/pharmacology , Zebrafish/metabolism
7.
Oncogene ; 40(21): 3748-3765, 2021 05.
Article in English | MEDLINE | ID: mdl-33941855

ABSTRACT

Activation of the transforming growth factor ß (TGFß) pathway modulates the expression of genes involved in cell growth arrest, motility, and embryogenesis. An expression screen for long noncoding RNAs indicated that TGFß induced mir-100-let-7a-2-mir-125b-1 cluster host gene (MIR100HG) expression in diverse cancer types, thus confirming an earlier demonstration of TGFß-mediated transcriptional induction of MIR100HG in pancreatic adenocarcinoma. MIR100HG depletion attenuated TGFß signaling, expression of TGFß-target genes, and TGFß-mediated cell cycle arrest. Moreover, MIR100HG silencing inhibited both normal and cancer cell motility and enhanced the cytotoxicity of cytostatic drugs. MIR100HG overexpression had an inverse impact on TGFß signaling responses. Screening for downstream effectors of MIR100HG identified the ligand TGFß1. MIR100HG and TGFB1 mRNA formed ribonucleoprotein complexes with the RNA-binding protein HuR, promoting TGFß1 cytokine secretion. In addition, TGFß regulated let-7a-2-3p, miR-125b-5p, and miR-125b-1-3p expression, all encoded by MIR100HG intron-3. Certain intron-3 miRNAs may be involved in TGFß/SMAD-mediated responses (let-7a-2-3p) and others (miR-100, miR-125b) in resistance to cytotoxic drugs mediated by MIR100HG. In support of a model whereby TGFß induces MIR100HG, which then enhances TGFß1 secretion, analysis of human carcinomas showed that MIR100HG expression correlated with expression of TGFB1 and its downstream extracellular target TGFBI. Thus, MIR100HG controls the magnitude of TGFß signaling via TGFß1 autoinduction and secretion in carcinomas.


Subject(s)
MicroRNAs/metabolism , Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism , Autocrine Communication , Cell Line, Tumor , Cell Proliferation/physiology , Humans , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Transforming Growth Factor beta1/genetics
8.
Onco Targets Ther ; 14: 455-467, 2021.
Article in English | MEDLINE | ID: mdl-33488100

ABSTRACT

INTRODUCTION: Multiple myeloma (MM) remains an incurable disease, and patient survival requires a better understanding of this malignancy's molecular aspects. Heparanase (HPSE) is highly expressed in aggressive MM cells and related to tumor growth, metastasis, and bortezomib (BTZ) resistance. Thus, targeting HPSE seems to be a promising approach for MM treatment, and because microRNAs (miRNAs) have emerged as potential regulators of HPSE expression, the use of extracellular vesicles (EVs) can allow the efficient delivery of therapeutic miRNAs. METHODS: We used prediction algorithms to identify potential miRNAs that regulate negatively HPSE expression. RT-qPCR was performed to assess miRNAs and HPSE expression in MM lines (U266 and RPMI-8226). Synthetic miRNA mimics were electroporated in MM cells to understand the miRNA contribution in HPSE expression, glycosaminoglycans (GAGs) profile, cell proliferation, and cell death induced by BTZ. EVs derived from HEK293T cells were engineered with miRNAs to evaluate their therapeutic potential combined with BTZ. RESULTS: It revealed a direct association between BTZ sensitivity, HPSE, and miR-1252-5p expressions. Moreover, overexpression of miR-1252-5p significantly reduced HPSE expression and HPSE enzymatic activity in MM cells. The higher level of miR-1252-5p was correlated with a reduction of cell viability and higher sensitivity to BTZ. Further, EVs carrying miR-1252-5p increased MM cells' sensitivity to BTZ treatment. CONCLUSION: These results showed that miR-1252-5p could negatively regulate HPSE in MM, indicating the use of EVs carrying miR-1252-5p as a potential novel BTZ sensitization approach in MM cells.

9.
Curr Res Immunol ; 2: 175-183, 2021.
Article in English | MEDLINE | ID: mdl-35492395

ABSTRACT

The content of tumor-derived extracellular vesicles (EVs) can regulate the tumor microenvironment and functionally acts in favor of cancer aggressiveness. To better elucidate the role of EVs in the interplay between immune system and tumor microenvironment, the purpose of this study was to analyze the effect of head and neck squamous cells carcinoma (HNSCC)-derived EVs on the modulation of inflammasomes - mediators of pyroptosis and secretion of inflammatory factors by macrophages. Our results showed that macrophages treated with the Vesicular Secretome Fraction (VSF) isolated from patient-derived HNSCC presented a reduction in the secretion of mature IL-1ß and caspase-1 without affecting cell viability. An analysis of the protein content of HNSCC-derived VSF by antibody array revealed that some of the most expressed proteins share a correlation with Transforming Growth Factor-beta (TGF-ß) activity. Since TGF-ß is related to the inhibition of the NF-kB-related pathways, including those required for the priming phase of the inflammasomes, we sought to evalute the interference of the VSF in the induction of inflammasome components. In fact, HNSCC-derived VSF inhibited the induction of pro-IL-1ß and pro-caspase-1 proteins and NLRP3 gene expression during the priming phase of inflammasome activation. Thus, our findings contribute to a better understanding of how tumor-derived EVs modulate inflammatory response by demonstrating their role in inhibiting NLRP3 inflammasomes.

10.
Medicine (Baltimore) ; 99(43): e22890, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33120836

ABSTRACT

Breast cancer is the most common malignancy affecting women worldwide. The insulin-like growth factor 1 (IGF-1) gene encodes a protein responsible for a wide variety of physiological processes, including differentiation and cell proliferation. Despite several studies on tumor tissues, no study has evaluated IGF-1 expression in the peripheral blood of women with recurrent breast cancer.In this cross-sectional study, IGF-1 expression in the peripheral blood of 146 women with breast cancer treated approximately 5 years ago was quantified by quantitative reverse transcription polymerase chain. The women were divided into 2 groups: non-recurrence (n = 85) and recurrence (n = 61). Statistical analysis of the data was performed using ANOVA, Mann-Whitney, and Chi-squared tests (P < .05).The results showed no significant difference in IGF-1 expression between the non-recurrence and recurrence groups (P = .988). In the subgroups of patients with lymph node involvement, no statistically significant difference was observed in IGF-1 expression between women with recurrence and those non-recurrence (P = .113). In patients without lymph node metastases, IGF-1 messenger ribonucleic acid (mRNA) expression levels were significantly higher in the non-recurrence group than in the recurrence group (P = .019). Furthermore, using the median IGF-1 mRNA expression as the cutoff point, it was obtained a statistically significant difference in tumor histological grade among women with recurrent breast cancer (P = .042).These data showed significantly higher IGF-1 expression in women without lymph node metastases in the non-recurrence group compared with the recurrence group. In addition, a significant difference was observed in median IGF-1 mRNA expression in relation to tumor histological grade in women with recurrent breast cancer.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/genetics , Insulin-Like Growth Factor I/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Aged, 80 and over , Brazil/epidemiology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cross-Sectional Studies , Female , Gene Expression/genetics , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoplasm Grading/methods , RNA, Messenger/genetics
11.
Tissue Cell ; 67: 101412, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32866727

ABSTRACT

Scaffolds composed of extracellular matrix (ECM) can assist tissue remodeling and repair following injury. The ECM is a complex biomaterial composed of proteins, glycoproteins, proteoglycans, and glycosaminoglycans, secreted by cells. The ECM contains fundamental biological cues that modulate cell behavior and serves as a structural scaffold for cell adhesion and growth. For clinical applications, where immune rejection is a constraint, ECM can be processed using decellularization methods intended to remove cells and donor antigens from tissue or organs, while preserving native biological cues essential for cell growth and differentiation. Recent studies show bioengineered organs composed by a combination of a diversity of materials and stem cells as a possibility of new therapeutic strategies to treat diseases that affect different tissues and organs, including the central nervous system (CNS). Nevertheless, the methodologies currently described for brain decellularization involve the use of several chemical reagents with many steps that ultimately limit the process of organ or tissue recellularization. Here, we describe for the first time a fast and straightforward method for complete decellularization of mice brain by the combination of rapid freezing and thawing following the use of only one detergent (Sodium dodecyl sulfate (SDS)). Our data show that using the protocol we describe here, the brain was entirely decellularized, while still maintaining ECM components that are essential for cell survival on the scaffold. Our results also show the cell-loading of the decellularized brain matrix with Neuro2a cells, which were identified by immunohistochemistry in their undifferentiated form. We conclude that this novel and simple method for brain decellularization can be used as a scaffold for cell-loading.


Subject(s)
Brain/physiology , Tissue Scaffolds/chemistry , Animals , Cell Differentiation , Cell Line , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/metabolism , Mice, Inbred C57BL , Nucleic Acids/metabolism , Sodium Dodecyl Sulfate
12.
Invest New Drugs ; 38(1): 70-78, 2020 02.
Article in English | MEDLINE | ID: mdl-31102120

ABSTRACT

Purpose Among alkaloids, abundant secondary metabolites in plants, aporphines constitute a class of compounds with interesting biological activities, including anticancer effects. The present study evaluated the anticancer activities of 14 substances, including four aporphine derivatives acquired through the biomonitoring of (±)-apomorphine hydrochloride total synthesis from 2-phenethylamine and 3,4-dimethoxybenzaldehyde against head and neck squamous cell carcinoma (HNSCC). Methods The cytotoxic effects of compounds against a panel of HNSCC cell lines were determined by PrestoBlue cell viability assay, while the genotoxicity of substances was evaluated by micronucleus test. Cell death was detected by flow cytometry (Annexin V/7AAD) and western blot analysis was used to detect the presence of cleaved Caspase-3 molecules. Results The aporphine and isoquinoline derivatives APO, C1, and A5 significantly reduced HNSCC cell viability and promoted DNA damages in these cells. Further, by activating the Caspase-3 pathway, these substances were able to induce apoptosis. Conclusion Our results revealed that APO, C1, and A5 exhibit cytotoxic effects in HNSCC cells. The mechanisms of action appear to be partly via the generation of DNA damages and apoptosis induction through Caspase-3 pathway activation. This study provides preclinical data that suggest a potential therapeutic role for APO, C1, and A5 against head and neck cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Aporphines/chemistry , Head and Neck Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis , Aporphines/pharmacology , Caspase 3/metabolism , Cell Proliferation , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Cells, Cultured
13.
Carcinogenesis ; 40(12): 1452-1461, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31436806

ABSTRACT

Management of locally advanced head and neck squamous cell carcinoma (HNSCC) requires a multi-prong approach comprising surgery, radiation and/or chemotherapy, yet outcomes are limited. This is largely due to a paucity of biomarkers that can predict response to specific treatment modalities. Here, we evaluated TGFß3 protein levels in extracellular vesicles (EVs) released by HNSCC cells as a predictor for response to chemoradiation therapy (CRT). To this end, specific EV-fractions were isolated from cell lines or HNSCC patient plasma, and TGFß3 protein was quantified. In patients treated with CRT, TGFß3 levels were found to be significantly higher in plasma EV-fractions or non-responders compared with responders. High levels of TGFß3 levels in Annexin V-EVs were associated with the worst progression-free survival. In vitro experiments demonstrated that TGFß3 silencing sensitized HNSCC cells to cytotoxic therapies, and this phenotype could be rescued by treatment with exogenous. In addition, specific EV-fractions shed by cisplatin-resistant cells were sufficient to transfer the resistant phenotype to sensitive cells through activation of TGFß-signaling pathway. Therefore, our data show that TGFß3 transmitted through EV plays a significant role in response to cytotoxic therapy, which can be exploited as a potential biomarker for CRT response in HNSCC patients treated with curative intent.


Subject(s)
Biomarkers, Tumor/blood , Extracellular Vesicles/metabolism , Head and Neck Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Transforming Growth Factor beta3/blood , Adult , Aged , Chemoradiotherapy/methods , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/physiology , Female , Head and Neck Neoplasms/blood , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Radiation Tolerance/physiology , Squamous Cell Carcinoma of Head and Neck/blood
14.
Mol Med Rep ; 19(6): 5023-5029, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31059005

ABSTRACT

Multiple myeloma (MM) is an incurable disease; a better understanding of the molecular aspects of this hematological malignancy could contribute to the development of new treatment strategies and help to improve the survival rates of patients with MM. Previously, the methylation status of the deleted in colorectal cancer (DCC) gene was correlated with the survival rate of patients with MM, thus the main goal of this study was to understand DCC contribution to MM tumorigenesis, and to assess the impact of DCC inhibition in the MM response to treatment with bortezomib. Our results demonstrated that hypermethylation of the DCC promoter inhibits gene expression, and DCC silencing is significantly correlated with a reduction in cell viability and an increase in cell death induced by bortezomib. In conclusion, our results suggested that hypermethylation is an important mechanism of DCC expression regulation in MM and that the absence of DCC contributes to the enhanced sensitivity to treatment with bortezomib.


Subject(s)
Bortezomib/pharmacology , DCC Receptor/metabolism , Down-Regulation/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DCC Receptor/antagonists & inhibitors , DCC Receptor/genetics , DNA Methylation/drug effects , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism
15.
BMC Cancer ; 19(1): 373, 2019 Apr 23.
Article in English | MEDLINE | ID: mdl-31014274

ABSTRACT

BACKGROUND: There is a paucity of plasma-based biomarkers that prospectively segregate the outcome of patients with head and neck squamous-cell carcinoma (HNSCC) treated with chemoradiation therapy (CRT). Plasma extracellular vesicles (EVs) might be an alternative source for discovery of new specific markers present in patients with HNSCC, which could help to re-direct patients to appropriate curative therapies without delay. METHODS: In order to identify new markers in plasma compartments, Cholerae toxin B chain (CTB) and Annexin V (AV) were used to isolate EVs from pooled plasma samples from patients with locally advanced HNSCC who responded (CR, n = 6) or presented incomplete response (NR, n = 6) to CRT. The crude plasma and EVs cargo were screened by antibody array. RESULTS: Of the 370 polypeptides detected, 119 proteins were specific to NR patients while 38 were exclusive of the CR subjects. The Gene Set Enrichment Analysis (GSEA) and Search Tool for the Retrieval of Interacting Genes (STRING) database analysis indicated that the content of circulating plasma EVs might have a relevant function for the tumor intercellular communication in the HNSCC patients. CONCLUSION: This study provides a list of potential markers present in plasma compartments that might contribute to the development of tools for prediction and assessment of CRT response and potentially guide therapeutic decisions in this context.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/pathology , Chemoradiotherapy/methods , Extracellular Vesicles/metabolism , Head and Neck Neoplasms/pathology , Adult , Aged , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/therapy , Clinical Trials, Phase II as Topic , Extracellular Vesicles/pathology , Female , Follow-Up Studies , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/therapy , Humans , Male , Middle Aged , Prognosis , Protein Array Analysis , Protein Interaction Maps
16.
Head Neck ; 41(5): 1199-1205, 2019 05.
Article in English | MEDLINE | ID: mdl-30803092

ABSTRACT

BACKGROUND: There is a paucity of plasma-based biomarkers that predict outcome in patients with head and neck squamous cell carcinoma (HNSCC) treated with chemoradiation therapy (CRT). Here, we evaluate the prognostic potential of plasma Melanoma-Antigen Recognized by T-cells 1 (MLANA) in this setting. METHODS: MLANA expression in HNSCC lines were evaluated by reverse transcription polymerase chain reaction, whereas plasma levels were quantified using ELISA in 48 patients with locally advanced HNSCC undergoing a phase 2 trial with CRT. RESULTS: MLANA is expressed at variable levels in a panel of HNSCC lines. In plasma, levels were elevated in patients with tumor relapse compared to those without (P < .004); 73.9% of the patients expressing high plasma MLANA levels progressed with recurrent disease (P = .020). Multivariate analysis showed that plasma MLANA levels and tumor resectability were independent prognostic factors for progression free survival. CONCLUSION: Plasma MLANA expression appears to be an effective noninvasive biomarker for outcomes in patients treated with CRT, and could potentially guide therapeutic decisions in this context.


Subject(s)
Chemoradiotherapy , Head and Neck Neoplasms/blood , MART-1 Antigen/blood , Squamous Cell Carcinoma of Head and Neck/blood , Adult , Aged , Biomarkers, Tumor/blood , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Induction Chemotherapy , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/therapy
17.
J Mol Neurosci ; 66(3): 383-389, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30284228

ABSTRACT

Glioblastoma (GBM) is an incurable disease ranked among the deadliest solid cancers worldwide. A better understanding on the molecular aspects of this malignancy could contribute to the development of new treatment strategies and help to improve survival rates. Previously, our group had shown that GBM patients expressing the cancer/testis antigen Opa Interacting Protein 5 (OIP5) present a longer survival period than the OIP5-negative group. The main goal of this study was to evaluate the OIP5 contribution to GBM tumorigenesis and assess the role of OIP5 in GBM cell response to lomustine, an alkylating agent used in the treatment of this malignancy. So, the effect of OIP5 knockdown was evaluated in A172 and T98G GBM cell lines. Our results demonstrated that downregulation of the OIP5 stimulates glioma cell viability and inhibits cell death-induced necrosis prompted by lomustine. In conclusion, our data shows that OIP5 expression in GBM cells seems to be able to enhance lomustine cytotoxic effects, reinforcing that this gene is a potential therapeutic target and putative molecular biomarker for treatment response in GBM.


Subject(s)
Brain Neoplasms/metabolism , Chromosomal Proteins, Non-Histone/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/genetics , Cell Cycle Proteins , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Humans , Lomustine/pharmacology
18.
Biomed Environ Sci ; 25(2): 245-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22998834

ABSTRACT

OBJECTIVE: This paper aims to evaluate the genotoxicity in peripheral blood lymphocytes of rats after exposure to sunlight at different time points of day in a tropical region of Brazil (5 degrees S, 42 degrees W). MATERIALS AND METHODS: Thirty Wistar-Hannover rats, three months old, were randomly divided into three groups of 10 animals each: Group I [control, without exposure to ultraviolet (UV) radiation], Group II (exposed to sunlight during 08:00 a.m. to 10:00 a.m.), and Group III (exposed to sunlight during 10:00 a.m. to 12:00 a.m.). After a week of exposure, peripheral blood samples were taken from the tail of these animals to prepare smears on two slides per animal. In 24 h after exposure to sunlight in Group III, a new collection was obtained to observe the repair activity. The alkaline comet assay was used in this study to evaluate the genotoxic activity of sunlight (P < 0.05). RESULTS: There was no statistical difference between Group I and II (P = 0.672). On the other hand, the exposure to sunlight in Group III showed genotoxic action in comparison to the other groups (P < 0.0001). Also, there was no significant repair in Group III R (P = 0.407). CONCLUSION: This study has shown a genotoxic potential of sunlight (UVA-B) in lymphocytes of mammals from 10:00 a.m. to 12:00 a.m., due to a higher intensity of UV in this tropical region.


Subject(s)
DNA Damage , Lymphocytes/radiation effects , Sunlight , Animals , Comet Assay , Rats , Rats, Wistar
19.
Int J Gynecol Cancer ; 22(1): 30-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21892088

ABSTRACT

OBJECTIVE: The objective of the study was to compare the effect of tamoxifen and raloxifene on the endometrium of female rats in persistent estrus, by Ki-67 protein expression. METHODS: The study comprised 60 Wistar-Hannover female rats in persistent estrus, induced by a single subcutaneous dose of 1.25 mg of testosterone propionate on the second day of age. At 90 days of life, the animals were randomly divided into 3 groups of 20 animals each. Group 1 (control), received only placebo; group 2, the animals were treated with tamoxifen, 250 µg/d; and group 3, the rats were treated with 750 µg/d of raloxifene by gavage during 30 days. Then, the animals were killed, and the endometrium was removed for immunohistochemical analysis of Ki-67 antigen expression. Statistical analysis was performed by ß regression model (P < 0.05). RESULTS: Mean percentages of Ki-67 protein expression in the endometrium of rats in persistent estrus were 43.21% ± 3.39%, 7.36% ± 0.95%, and 7.20% ± 0.76% in groups 1, 2 and 3, respectively (P < 0.001). There was no statistical difference between groups 2 and 3 (P = 0.7159). CONCLUSIONS: The present results indicate that, at the doses and during the time of treatment used, both tamoxifen and raloxifene induce atrophy in a similar way of endometrial epithelium of rats in persistent estrus.


Subject(s)
Cell Proliferation/drug effects , Endometrium/drug effects , Estrus , Ki-67 Antigen/metabolism , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Animals , Endometrium/cytology , Endometrium/metabolism , Female , Immunohistochemistry , Random Allocation , Rats , Regression Analysis , Single-Blind Method
SELECTION OF CITATIONS
SEARCH DETAIL
...