Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Pers Med ; 11(2)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572343

ABSTRACT

This study represents a novel proof of concept of the clinical utility of miRNAs from exhaled breath condensate (EBC) as biomarkers of lung cancer (LC). Genome-wide miRNA profiling and machine learning analysis were performed on EBC from 21 healthy volunteers and 21 LC patients. The levels of 12 miRNAs were significantly altered in EBC from LC patients where a specific signature of miR-4507, miR-6777-5p and miR-451a distinguished these patients with high accuracy. Besides, a distinctive miRNA profile between LC adenocarcinoma and squamous cell carcinoma was observed, where a combined panel of miR-4529-3p, miR-8075 and miR-7704 enabling discrimination between them. EBC levels of miR-6777-5p, 6780a-5p and miR-877-5p predicted clinical outcome at 500 days. Two additional miRNA signatures were also associated with other clinical features such as stage and invasion status. Dysregulated EBC miRNAs showed potential target genes related to LC pathogenesis, including CDKN2B, PTEN, TP53, BCL2, KRAS and EGFR. We conclude that EBC miRNAs might allow the identification, stratification and monitorization of LC, which could lead to the development of precision medicine in this and other respiratory diseases.

2.
Lab Invest ; 101(3): 292-303, 2021 03.
Article in English | MEDLINE | ID: mdl-33262438

ABSTRACT

Cancer stem cells (CSCs) are involved in the resistance of estrogen (ER)-positive breast tumors against endocrine therapy. On the other hand, nitric oxide (NO) plays a relevant role in CSC biology, although there are no studies addressing how this important signaling molecule may contribute to resistance to antihormonal therapy in ER+ breast cancer. Therefore, we explored whether targeting NO in ER+ breast cancer cells impacts CSC subpopulation and sensitivity to hormonal therapy with tamoxifen. NO was targeted in ER+ breast cancer cells by specific NO depletion and NOS2 silencing and mammosphere formation capacity, stem cell markers and tamoxifen sensitivity were analyzed. An orthotopic breast tumor model in mice was also performed to analyze the efficacy of NO-targeted therapy plus tamoxifen. Kaplan-Meier curves were made to analyze the association of NOS2 gene expression with survival of ER+ breast cancer patients treated with tamoxifen. Our results show that targeting NO inhibited mamosphere formation, CSC markers expression and increased the antitumoral efficacy of tamoxifen in ER+ breast cancer cells, whereas tamoxifen-resistant cells displayed higher expression levels of NOS2 and Notch-1 compared with parental cells. Notably, NO-targeted therapy plus tamoxifen was more effective than either treatment alone in an orthotopic breast tumor model in immunodeficient mice. Furthermore, low NOS2 expression was significantly associated with a higher metastasis-free survival in ER+ breast cancer patients treated with tamoxifen. In conclusion, our data support that NO-targeted therapy in ER+ breast cancer may contribute to increase the efficacy of antihormonal therapy avoiding the development of resistance to these treatments.


Subject(s)
Antineoplastic Agents, Hormonal , Breast Neoplasms , Nitric Oxide , Receptors, Estrogen/metabolism , Tamoxifen , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Silencing , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Signal Transduction/drug effects , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
3.
Biochem Pharmacol ; 176: 113769, 2020 06.
Article in English | MEDLINE | ID: mdl-31862448

ABSTRACT

Nitric oxide (NO) has been highlighted as an important agent in tumor processes. However, a complete understanding of the mechanisms by which this simple diatomic molecule contributes in tumorigenesis is lacking. Evidence is rapidly accumulating that metabolic reprogramming is a major new aspect of NO biology and this review is aimed to summarize recent research progress on this novel feature that expands the complex and multifaceted role of NO in cancer. Therefore, we discuss how NO may influence glucose and glutamine utilization by tumor cells, and its participation in the regulation of mitochondrial function and dynamics, that is an important mechanism through which cancer cells reprogram their metabolism to meet the biosynthetic needs of rapid proliferation. Finally, we also discuss the NO-related metabolic rewiring involved in the modification of the tumor microenvironment to support cancer invasion and the escape from immune system-mediated recognition. Protein S-nitrosylation appears as a common mechanism by which NO signaling reprograms metabolism. Hence, future research is needed on dysregulated S-nitrosylation/denitrosylation in cancer to comprehend the NO-induced metabolic changes in tumor cells and the role of NO in the metabolic crosstalk within tumor microenvironment.


Subject(s)
Glucose/metabolism , Glutamine/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Nitric Oxide/metabolism , Cell Proliferation , Energy Metabolism , Humans , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment
4.
J Cell Mol Med ; 23(12): 8219-8232, 2019 12.
Article in English | MEDLINE | ID: mdl-31560832

ABSTRACT

Newly emerged proteomic methodologies, particularly data-independent acquisition (DIA) analysis-related approaches, would improve current gene expression-based classifications of colorectal cancer (CRC). Therefore, this study was aimed to identify protein expression signatures using SWATH-MS DIA and targeted data extraction, to aid in the classification of molecular subtypes of CRC and advance in the diagnosis and development of new drugs. For this purpose, 40 human CRC samples and 7 samples of healthy tissue were subjected to proteomic and bioinformatic analysis. The proteomic analysis identified three different molecular CRC subtypes: P1, P2 and P3. Significantly, P3 subtype showed high agreement with the mesenchymal/stem-like subtype defined by gene expression signatures and characterized by poor prognosis and survival. The P3 subtype was characterized by decreased expression of ribosomal proteins, the spliceosome, and histone deacetylase 2, as well as increased expression of osteopontin, SERPINA 1 and SERPINA 3, and proteins involved in wound healing, acute inflammation and complement pathway. This was also confirmed by immunodetection and gene expression analyses. Our results show that these tumours are characterized by altered expression of proteins involved in biological processes associated with immune evasion and metastasis, suggesting new therapeutic options in the treatment of this aggressive type of CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Proteome/metabolism , Proteomics/methods , Aged , Biomarkers, Tumor/genetics , Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Gene Regulatory Networks , Humans , Immune Evasion , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Proteome/genetics
5.
BMC Biol ; 16(1): 3, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29329541

ABSTRACT

BACKGROUND: Nitric oxide (NO) has been highlighted as an important agent in cancer-related events. Although the inducible nitric oxide synthase (iNOS) isoform has received most attention, recent studies in the literature indicate that the endothelial isoenzyme (eNOS) can also modulate different tumor processes including resistance, angiogenesis, invasion, and metastasis. However, the role of eNOS in cancer stem cell (CSC) biology and mesenchymal tumors is unknown. RESULTS: Here, we show that eNOS was significantly upregulated in VilCre ERT2 Apc fl/+ and VilCre ERT2 Apc fl/fl mouse intestinal tissue, with intense immunostaining in hyperproliferative crypts. Similarly, the more invasive VilCre ERT2 Apc fl/+ Pten fl/+ mouse model showed an overexpression of eNOS in intestinal tumors whereas this isoform was not expressed in normal tissue. However, none of the three models showed iNOS expression. Notably, when 40 human colorectal tumors were classified into different clinically relevant molecular subtypes, high eNOS expression was found in the poor relapse-free and overall survival mesenchymal subtype, whereas iNOS was absent. Furthermore, Apc fl/fl organoids overexpressed eNOS compared with wild-type organoids and NO depletion with the scavenger carboxy-PTIO (c-PTIO) decreased the proliferation and the expression of stem-cell markers, such as Lgr5, Troy, Vav3, and Slc14a1, in these intestinal organoids. Moreover, specific NO depletion also decreased the expression of CSC-related proteins in human colorectal cancer cells such as ß-catenin and Bmi1, impairing the CSC phenotype. To rule out the contribution of iNOS in this effect, we established an iNOS-knockdown colorectal cancer cell line. NO-depleted cells showed a decreased capacity to form tumors and c-PTIO treatment in vivo showed an antitumoral effect in a xenograft mouse model. CONCLUSION: Our data support that eNOS upregulation occurs after Apc loss, emerging as an unexpected potential new target in poor-prognosis mesenchymal colorectal tumors, where NO scavenging could represent an interesting therapeutic alternative to targeting the CSC subpopulation.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cell Proliferation/physiology , Colorectal Neoplasms/enzymology , Intestines/enzymology , Mesenchymal Stem Cells/enzymology , Nitric Oxide Synthase Type III/physiology , Animals , Caco-2 Cells , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Intestines/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays/methods
6.
Oncotarget ; 8(46): 81462-81474, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-29113405

ABSTRACT

The association between breast cancer (BCa) presence and altered glucose/insulin metabolism is controversial likely due to an inaccurate insulin resistance (IR) assessment and inappropriate stratification of patients by body-mass index (BMI) and menopausal state. 148 women with suspect of sporadic BCa were stratified by BMI and menopause. Fasting levels of glucose, insulin, glycohemoglobin and selected IR-related and tumor-derived markers were measured. Glucose/insulin levels during OGTT were used to calculate insulin resistance/sensitivity indexes. Analysis of 77 BCa-bearing patients and 71 controls showed an association between BCa and IR as demonstrated by impaired glucose/insulin homeostasis (increased fasting- and OGTT-induced glucose levels) and deteriorated IR indexes, which was especially patent in premenopausal women. The association between BCa presence and IR was markedly influenced by BMI, being obese BCa patients significantly more insulin resistant than controls. BCa presence was associated to elevated levels of IR (glucose, triglycerides) and tumor-derived (VEGF) markers, especially in overweight/obese patients. BCa presence is associated to IR in overweight/obese premenopausal but not in premenopausal normal weight or postmenopausal women. Our data support a bidirectional relationship between dysregulated/imbalanced glucose/insulin metabolism and BCa, as tumor- and IR-markers are correlated with the impairment of glucose/insulin metabolism in overweight/obese premenopausal BCa patients.

7.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L664-L676, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28619761

ABSTRACT

We explored whether the proteomic analysis of exhaled breath condensate (EBC) may provide biomarkers for noninvasive screening for the early detection of lung cancer (LC). EBC was collected from 192 individuals [49 control (C), 49 risk factor-smoking (S), 46 chronic obstructive pulmonary disease (COPD) and 48 LC]. With the use of liquid chromatography and tandem mass spectrometry, 348 different proteins with a different pattern among the four groups were identified in EBC samples. Significantly more proteins were identified in the EBC from LC compared with other groups (C: 12.4 ± 1.3; S: 15.3 ± 1; COPD: 14 ± 1.6; LC: 24.2 ± 3.6; P = 0.0001). Furthermore, the average number of proteins identified per sample was significantly higher in LC patients, and receiver operating characteristic curve (ROC) analysis showed an area under the curve of 0.8, indicating diagnostic value. Proteins frequently detected in EBC, such as dermcidin and hornerin, along with others much less frequently detected, such as hemoglobin and histones, were identified. Cytokeratins (KRTs) were the most abundant proteins in EBC samples, and levels of KRT6A, KRT6B, and KRT6C isoforms were significantly higher in samples from LC patients (P = 0.0031, 0.0011, and 0.0009, respectively). Moreover, the amount of most KRTs in EBC samples from LC patients showed a significant positive correlation with tumor size. Finally, we used a random forest algorithm to generate a robust model using EBC protein data for the diagnosis of patients with LC where the area under the ROC curve obtained indicated a good classification (82%). Thus this study demonstrates that the proteomic analysis of EBC samples is an appropriated approach to develop biomarkers for the diagnosis of lung cancer.


Subject(s)
Biomarkers/metabolism , Breath Tests/methods , Early Detection of Cancer/methods , Lung Neoplasms/diagnosis , Proteome/metabolism , Small Cell Lung Carcinoma/diagnosis , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Breath Tests/instrumentation , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Exhalation , Female , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Proteomics/methods , Small Cell Lung Carcinoma/metabolism
8.
Oncotarget ; 8(13): 21754-21769, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423516

ABSTRACT

Here we showed that the addition of the COX-2 inhibitor celecoxib improved the antitumor efficacy in colorectal cancer (CRC) of the monoclonal anti-EGFR antibody cetuximab. The addition of celecoxib augmented the efficacy of cetuximab to inhibit cell proliferation and to induce apoptosis in CRC cells. Moreover, the combination of celecoxib and cetuximab was more effective than either treatment alone in reducing the tumor volume in a mouse xenograft model. The combined treatment enhanced the inhibition of EGFR signaling and altered the subcellular distribution of ß-catenin. Moreover, knockdown of FOXM1 showed that this transcription factor participates in this enhanced antitumoral response. Besides, the combined treatment decreased ß-catenin/FOXM1 interaction and reduced the cancer stem cell subpopulation in CRC cells, as indicated their diminished capacity to form colonospheres. Notably, the inmunodetection of FOXM1 in the nuclei of tumor cells in human colorectal adenocarcinomas was significantly associated with response of patients to cetuximab. In summary, our study shows that the addition of celecoxib enhances the antitumor efficacy of cetuximab in CRC due to impairment of EGFR-RAS-FOXM1-ß-catenin signaling axis. Results also support that FOXM1 could be a predictive marker of response of mCRC patients to cetuximab therapy.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Celecoxib/pharmacology , Cetuximab/pharmacology , Colorectal Neoplasms/pathology , Signal Transduction/drug effects , Animals , Blotting, Western , Drug Synergism , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Forkhead Box Protein M1/drug effects , Forkhead Box Protein M1/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Confocal , Xenograft Model Antitumor Assays , beta Catenin/drug effects , beta Catenin/metabolism , ras Proteins/drug effects , ras Proteins/metabolism
9.
Sci Rep ; 7: 42002, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181484

ABSTRACT

Chimeric somatostatin/dopamine compounds such as BIM-23A760, an sst2/sst5/D2 receptors-agonist, have emerged as promising new approaches to treat pituitary adenomas. However, information on direct in vitro effects of BIM-23A760 in normal and tumoral pituitaries remains incomplete. The objective of this study was to analyze BIM-23A760 effects on functional parameters (Ca2+ signaling, hormone expression/secretion, cell viability and apoptosis) in pituitary adenomas (n = 74), and to compare with the responses of normal primate and human pituitaries (n = 3-5). Primate and human normal pituitaries exhibited similar sst2/sst5/D2 expression patterns, wherein BIM-23A760 inhibited the expression/secretion of several pituitary hormones (specially GH/PRL), which was accompanied by increased sst2/sst5/D2 expression in primates and decreased Ca2+ concentration in human cells. In tumoral pituitaries, BIM-23A760 also inhibited Ca2+ concentration, hormone secretion/expression and proliferation. However, BIM-23A760 elicited stimulatory effects in a subset of GHomas, ACTHomas and NFPAs in terms of Ca2+ signaling and/or hormone secretion, which was associated with the relative somatostatin/dopamine-receptors levels, especially sst5 and sst5TMD4. The chimeric sst2/sst5/D2 compound BIM-23A760 affects multiple, clinically relevant parameters on pituitary adenomas and may represent a valuable therapeutic tool. The relative ssts/D2 expression profile, particularly sst5 and/or sst5TMD4 levels, might represent useful molecular markers to predict the ultimate response of pituitary adenomas to BIM-23A760.


Subject(s)
Adenoma/metabolism , Dopamine Agonists/pharmacology , Dopamine/analogs & derivatives , Pituitary Gland/drug effects , Pituitary Neoplasms/metabolism , Somatostatin/analogs & derivatives , Adolescent , Adult , Aged , Animals , Apoptosis , Calcium Signaling , Cell Survival , Cells, Cultured , Dopamine/pharmacology , Exocytosis , Female , Humans , Male , Middle Aged , Papio , Pituitary Gland/cytology , Pituitary Gland/metabolism , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology
10.
Biochim Biophys Acta ; 1862(4): 601-610, 2016 04.
Article in English | MEDLINE | ID: mdl-26854735

ABSTRACT

The monoclonal antibody trastuzumab against HER2/neu, which is overexpressed in 15-20% of breast cancers, has clinical efficacy but many patients do not respond to initial treatment or develop resistance during treatment. Nitric oxide (NO) regulates cell signaling by targeting specific cysteine residues in proteins, forming S-nitrosothiols (SNO) in a process known as S-nitrosylation. We previously reported that molecular characteristics in breast cancer may dictate the tumor response to impaired SNO homeostasis. In the present study, we explored the role of SNO homeostasis in HER2 breast tumors. The antiproliferative action of trastuzumab in HER2-overexpressing BT-474 and SKBR-3 cells was suppressed when S-nitrosoglutathione reductase (GSNOR/ADH5) activity, which plays a key role in SNO homeostasis, was specifically inhibited with the pyrrole derivative compound N6022. Moreover, GSNOR inhibition restored the activation of survival signaling pathways involved in the resistance to anti-HER2 therapies (AKT, Src and c-Abl kinases and TrkA/NRTK1, TrkB/NRTK2, EphA1 and EphA3 receptors) and reduced the apoptotic effect of trastuzumab. Accordingly, GSNOR inhibition augmented the S-nitrosylation of apoptosis-related proteins, including Apaf-1, pSer73/63 c-Jun, calcineurin subunit α and HSF1. In agreement with in vitro data, immunohistochemical analyses of 51 breast tumors showed that HER2 expression was associated with lower expression of GSNOR protein. Moreover, gene expression analysis confirmed that high ADH5/GSNOR gene expression was associated with high patient survival rates in HER2 tumors. In conclusion, our data provide evidence of molecular mechanisms contributing to the progression of HER2+ breast cancers and could facilitate the development of therapeutic options to counteract resistance to anti-HER2 therapies.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm/drug effects , Homeostasis/drug effects , Receptor, ErbB-2/metabolism , S-Nitrosothiols/metabolism , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Female , Humans , MCF-7 Cells
11.
Eur J Clin Invest ; 45(12): 1325-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26509357

ABSTRACT

BACKGROUND: Currently, there are no predictive biomarkers for anti-angiogenic strategies in cancer, but response to anti-angiogenic drugs is associated with development of hypertension secondary to treatment. Therefore, this study explored the clinical relevance of genetic polymorphisms in some components of the renin-angiotensin system (RAS). MATERIAL AND METHODS: Genomic DNA was isolated from peripheral blood from 95 metastatic breast or colorectal cancer patients treated with bevacizumab, and AGTR1-A1166C (rs5186), AGT-M235T (rs699) SNPs and ACE I/D (rs4646994) polymorphisms were genotyped using RT-PCR. Circulating vascular endothelial grow factor and angiotensin converting enzyme (ACE) levels were analysed using ELISA kits. The antitumoral activity of bevacizumab was assayed in mice orthotopically xenografted with AGTR1-overexpressing breast cancer cells. RESULTS: The ACE IN/IN genotype was associated with a higher rate of disease progression compared to DEL/IN and DEL/DEL genotypes (36% vs. 11·1% P < 0·05). Similarly, AGTR1-1166A/A genotype was also associated with a higher rate of disease progression compared to AGTR1-1166A/C and AGTR1-1166C/C genotypes (24·4% vs. 2·7% P < 0·01). ACE IN/IN genotype was also found to be associated with shorter time to treatment failure compared to ACE IN/DEL and ACE DEL/DEL genotypes (14 weeks vs. 41·71, P = 0·033), whereas circulating ACE levels were found to be associated with a better response to bevacizumab treatment. Besides, in vivo experiments showed a significantly higher antitumoral activity of bevacizumab in tumours derived from AGTR1-overexpressing breast cancer cells. CONCLUSIONS: A higher activity of ACE-angiotensin-II-AGTR1 axis is associated with a better response to bevacizumab, supporting that the RAS can be an important source of potential predictive markers of response to anti-angiogenic drugs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Breast Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Renin-Angiotensin System/genetics , Adult , Aged , Aged, 80 and over , Angiotensin II/metabolism , Animals , Breast Neoplasms/genetics , Colorectal Neoplasms/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Heterografts/metabolism , Humans , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Metastasis , Neoplasm Transplantation , Peptidyl-Dipeptidase A/metabolism , Polymorphism, Genetic , Prospective Studies , Receptor, Angiotensin, Type 1/genetics , Retrospective Studies , Vascular Endothelial Growth Factor A/metabolism
12.
PLoS One ; 10(6): e0131363, 2015.
Article in English | MEDLINE | ID: mdl-26107817

ABSTRACT

Despite the demonstrated benefits of anti-EGFR/VEGF targeted therapies in metastatic colorectal cancer (mCRC), many patients initially respond, but then show evidence of disease progression. New therapeutic strategies are needed to make the action of available drugs more efficient. Our study aimed to explore whether simultaneous targeting of EGFR/VEGF and cyclooxygenase-2 (COX-2) may aid the treatment and management of mCRC patients. The dual tyrosine kinase inhibitor AEE788 and celecoxib were used to inhibit EGFR/VEGFR and COX-2, respectively, in colorectal cancer cells. COX-2 inhibition with celecoxib augmented the antitumoral and antiangiogenic efficacy of AEE788, as indicated by the inhibition of cell proliferation, induction of apoptosis and G1 cell cycle arrest, down-regulation of VEGF production by cancer cells and reduction of cell migration. These effects were related with a blockade in the EGFR/VEGFR signaling axis. Notably, the combined AEE788/celecoxib treatment prevented ß-catenin nuclear accumulation in tumor cells. This effect was associated with a significant downregulation of FOXM1 protein levels and an impairment in the interaction of this transcription factor with ß-catenin, which is required for its nuclear localization. Furthermore, the combined treatment also reduced the expression of the stem cell markers Oct 3/4, Nanog, Sox-2 and Snail in cancer cells, and contributed to the diminution of the CSC subpopulation, as indicated by colonosphere formation assays. In conclusion, the combined treatment of AEE788 and celecoxib not only demonstrated enhanced anti-tumoral efficacy in colorectal cancer cells, but also reduced colon CSCs subpopulation by targeting stemness-related pathways. Therefore, the simultaneous targeting of EGFR/VEGF and COX-2 may aid in blocking mCRC progression and improve the efficacy of existing therapies in colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2/chemistry , ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Apoptosis , Caco-2 Cells , Celecoxib/administration & dosage , Cell Cycle , Cell Proliferation , Disease Progression , Enzyme-Linked Immunosorbent Assay , G1 Phase , Gene Expression Regulation, Neoplastic , Genes, ras , HCT116 Cells , Humans , Microscopy, Confocal , Neovascularization, Pathologic , Purines/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
13.
PLoS One ; 9(6): e99143, 2014.
Article in English | MEDLINE | ID: mdl-24932611

ABSTRACT

The induction of polyploidy is considered the reproductive end of cells, but there is evidence that polyploid giant cancer cells (PGCCs) contribute to cell repopulation during tumor relapse. However, the role of these cells in the development, progression and response to therapy in colon cancer remains undefined. Therefore, the main objective of this study was to investigate the generation of PGCCs in colon cancer cells and identify mechanisms of formation. Treatment of HCT-116 and Caco-2 colon cancer cells with the hypoxia mimic CoCl2 induced the formation of cells with larger cell and nuclear size (PGCCs), while the cells with normal morphology were selectively eliminated. Cytometric analysis showed that CoCl2 treatment induced G2 cell cycle arrest and the generation of a polyploid cell subpopulation with increased cellular DNA content. Polyploidy of hypoxia-induced PGCCs was confirmed by FISH analysis. Furthermore, CoCl2 treatment effectively induced the stabilization of HIF-1α, the differential expression of a truncated form of p53 (p47) and decreased levels of cyclin D1, indicating molecular mechanisms associated with cell cycle arrest at G2. Generation of PGCCs also contributed to expansion of a cell subpopulation with cancer stem cells (CSCs) characteristics, as indicated by colonosphere formation assays, and enhanced chemoresistance to 5-fluorouracil and oxaliplatin. In conclusion, the pharmacological induction of hypoxia in colon cancer cells causes the formation of PGCCs, the expansion of a cell subpopulation with CSC characteristics and chemoresistance. The molecular mechanisms involved, including the stabilization of HIF-1 α, the involvement of p53/p47 isoform and cell cycle arrest at G2, suggest novel targets to prevent tumor relapse and treatment failure in colon cancer.


Subject(s)
Adenocarcinoma/pathology , Cell Hypoxia , Cobalt/pharmacology , Colonic Neoplasms/pathology , Giant Cells/drug effects , Neoplastic Stem Cells/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Cyclin D1/metabolism , DNA, Neoplasm/analysis , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , G2 Phase/drug effects , Giant Cells/ultrastructure , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Situ Hybridization, Fluorescence , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , Polyploidy , Protein Isoforms/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
14.
PLoS One ; 9(1): e85527, 2014.
Article in English | MEDLINE | ID: mdl-24465589

ABSTRACT

Somatostatin receptors (ssts) are expressed in thyroid cancer cells, but their biological significance is not well understood. The aim of this study was to assess ssts in well differentiated (WDTC) and poorly differentiated thyroid cancer (PDTC) by means of imaging and molecular tools and its relationship with the efficacy of somatostatin analog treatment. Thirty-nine cases of thyroid carcinoma were evaluated (20 PDTC and 19 WDTC). Depreotide scintigraphy and mRNA levels of sst-subtypes, including the truncated variant sst5TMD4, were carried out. Depreotide scans were positive in the recurrent tumor in the neck in 6 of 11 (54%) PDTC, and in those with lung metastases in 5/11 cases (45.4%); sst5TMD4 was present in 18/20 (90%) of PDTC, being the most densely expressed sst-subtype, with a 20-fold increase in relation to sst2. In WDTC, sst2 was the most represented, while sst5TMD4 was not found; sst2 was significantly increased in PDTC in comparison to WDTC. Five depreotide positive PDTC received octreotide for 3-6 months in a pilot study with no changes in the size of the lesions in 3 of them, and a significant increase in the pulmonary and cervical lesions in the other 2. All PDTC patients treated with octreotide showed high expression of sst5TMD4. ROC curve analysis demonstrated that only sst5TMD4 discriminates between PDTC and WDTC. We conclude that sst5TMD4 is overexpressed in PDTC and may be involved in the lack of response to somatostatin analogue treatment.


Subject(s)
Head and Neck Neoplasms/genetics , Lung Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , RNA, Messenger/genetics , Receptors, Somatostatin/genetics , Thyroid Neoplasms/genetics , Adult , Aged , Antineoplastic Agents, Hormonal/therapeutic use , Female , Gene Expression , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/secondary , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Octreotide/therapeutic use , Protein Isoforms/genetics , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology
15.
Mass Spectrom Rev ; 33(1): 7-20, 2014.
Article in English | MEDLINE | ID: mdl-23775552

ABSTRACT

Many of nitric oxide (NO) actions are mediated through the coupling of a nitroso moiety to a reactive cysteine leading to the formation of a S-nitrosothiol (SNO), a process known as S-nitrosylation or S-nitrosation. In many cases this reversible post-translational modification is accompanied by altered protein function and aberrant S-nitrosylation of proteins, caused by altered production of NO and/or impaired SNO homeostasis, has been repeatedly reported in a variety of pathophysiological settings. A growing number of studies are directed to the identification and characterization of those proteins that undergo S-nitrosylation and the analysis of S-nitrosoproteomes under pathological conditions is beginning to be reported. The study of these S-nitrosoproteomes has been fueled by advances in proteomic technologies that are providing researchers with improved tools for exploring this post-translational modification. Here we review novel refinements and improvements to these methods, and some recent studies of the S-nitrosoproteome in disease.


Subject(s)
Proteins/chemistry , Proteomics/methods , S-Nitrosothiols/analysis , Animals , Cysteine/analysis , Cysteine/metabolism , Humans , Nitric Oxide/metabolism , Nitrosation , Protein Processing, Post-Translational , Proteins/metabolism , S-Nitrosothiols/metabolism
16.
Breast Cancer Res ; 14(6): R153, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23216744

ABSTRACT

INTRODUCTION: Protein denitrosylation by thioredoxin reductase (TrxR) is key for maintaining S-nitrosothiol (SNO) homeostasis, although its role in tumor progression is unknown. Therefore, the present study aimed to assess the role of altered SNO homeostasis in breast cancer cells. METHODS: The impairment of SNO homeostasis in breast cancer cells was achieved with the highly specific TrxR inhibitor auranofin and/or exposure to S-nitroso-L-cysteine. S-nitrosylated proteins were detected using the biotin switch assay. Estrogen receptor (ER) alpha knockdown was achieved using RNA silencing technologies and subcellular localization of ERα was analyzed by confocal microscopy. The Oncomine database was explored for TrxR1 (TXNRD1) expression in breast tumors and TrxR1, ER and p53 expression was analyzed by immunohistochemistry in a panel of breast tumors. RESULTS: The impairment of SNO homeostasis enhanced cell proliferation and survival of ER+ MCF-7 cells, but not of MDA-MB-231 (ER-, mut p53) or BT-474 (ER+, mut p53) cells. This enhanced cell growth and survival was associated with Akt, Erk1/2 phosphorylation, and augmented cyclin D1 expression and was abolished by the ER antagonist fulvestrant or the p53 specific inhibitor pifithrin-α. The specific silencing of ERα expression in MCF-7 cells also abrogated the growth effect of TrxR inhibition. Estrogenic deprivation in MCF-7 cells potentiated the pro-proliferative effect of impaired SNO homeostasis. Moreover, the subcellular distribution of ERα was altered, with a predominant nuclear localization associated with phosphorylation at Thr311 in those cells with impaired SNO homeostasis. The impairment of SNO homeostasis also expanded a cancer stem cell-like subpopulation in MCF-7 cells, as indicated by the increase of percentage of CD44+ cells and the augmented capability to form mammospheres in vitro. Notably, ER+ status in breast tumors was significantly associated with lower TXNDR1 mRNA expression and immunohistochemical studies confirmed this association, particularly when p53 abnormalities were absent. CONCLUSION: The ER status in breast cancer may dictate tumor response to different nitrosative environments. Impairment of SNO homeostasis confers survival advantages to ER+ breast tumors, and these molecular mechanisms may also participate in the development of resistance against hormonal therapies that arise in this type of mammary tumors.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , S-Nitrosothiols/chemistry , Antirheumatic Agents/pharmacology , Auranofin/pharmacology , Benzothiazoles/pharmacology , Breast Neoplasms/drug therapy , CD24 Antigen/biosynthesis , Cell Proliferation , Cell Survival , Cyclin D1/biosynthesis , Cysteine/analogs & derivatives , Cysteine/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fulvestrant , Homeostasis , Humans , Hyaluronan Receptors/biosynthesis , MCF-7 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , S-Nitrosothiols/pharmacology , Spheroids, Cellular , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/biosynthesis , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/biosynthesis
17.
Expert Rev Proteomics ; 9(1): 59-69, 2012.
Article in English | MEDLINE | ID: mdl-22292824

ABSTRACT

Nitric oxide (NO) can modulate cell function by the coupling of a nitroso moiety to a reactive cysteine in target proteins leading to the formation of a S-nitrosothiol (SNO), a process commonly known as S-nitrosylation. Aberrant S-nitrosylation of proteins, caused by altered production of NO and/or impaired SNO homeostasis, constitutes a mechanism that has been recently postulated in numerous pathophysiological settings. The thiol microenvironment, cellular redox environment, and activity of transnitrosylases and denitrosylases have been proposed as determinant factors for the specificity of S-nitrosylation. A number of methodological approaches have recently been developed for the proteomic identification of S-nitrosylated proteins and/or the identification of specific sites of nitrosylation. This review will consider novel aspects of SNO homeostasis and S-nitrosylation, the latest proteomic methods for the identification of S-nitrosylated cysteines in proteins, and how these novel technologies will impact our current knowledge of the role of deregulated S-nitrosylation in disease.


Subject(s)
Disease , Nitroso Compounds/metabolism , Proteomics , Humans , Nitric Oxide/biosynthesis , Nitric Oxide/physiology
18.
World J Hepatol ; 2(3): 127-35, 2010 Mar 27.
Article in English | MEDLINE | ID: mdl-21160983

ABSTRACT

AIM: To identify new markers of hepatocellular carcinoma (HCC) using a proteomic analysis. METHODS: Patients with liver cirrhosis of the three most frequent etiologies: hepatitis C virus, hepatitis B virus and alcoholic liver disease, were included in the study. The samples were analysed by 2D-electrophoresis in order to determine the differential protein expression. The proteins were separated according to the charge in immobilized pH 3-10 gradient strips and then by sodium dodecyl sulfate polyacrylamide gel electrophoresis. Proteins of interest were excised, digested with trypsin and the resulting peptides were separated and identified. RESULTS: Three differentially expressed apolipoproteins (Apo) were identified based on the protein profile using proteomic techniques: Apo-A1, Apo-A4 and Apo-E. Apo-A4 levels were significantly lower in HCC than in non-HCC patients regardless of etiology (P < 0.01). Multivariate logistic regression showed that Apo-A4 and Apo-A1 were the only independent factors related to HCC diagnosis (P < 0.05). The receiver operating characteristic (ROC) curve including both Apo-A4 and Apo-A1 showed an area under the ROC of 0.944 (P < 0.001), a sensitivity of 0.89 and a specificity of 0.81 for diagnosis of HCC. CONCLUSION: Apo-A4 and Apo-A1 may be used clinically as biomarkers of HCC with a high sensibility and specificity. These findings may provide additional insights into the mechanism of HCC development and progression.

19.
Anticancer Res ; 30(2): 415-21, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20332448

ABSTRACT

BACKGROUND/AIM: S-Nitrosoglutathione reductase (GSNOR) and thioredoxin enzyme systems participate in cellular defence against nitrosative stress. Pharmacological interventions against these enzyme systems might represent valuable strategies to impair S-nitrosothiol (SNO) homeostasis in tumour cells. MATERIALS AND METHODS: Human HepG2 cells were pre-treated with mithramycin A or auranofin and exposed to S-nitroso-L-cysteine. GSNOR mRNA levels were analyzed by quantitative real-time reverse transcriptase-polymerase chain reaction and S-nitrosated proteins were detected and purified using the biotin-switch approach. Proteins were identified using electrospray ionization tandem mass spectrometry. RESULTS: Mithramycin interfered with GSNOR induction resulting in an increased cellular sensitivity to protein S-nitrosation. Moreover, the thioredoxin reductase inhibitor auranofin also increased cellular susceptibility to S-nitrosoprotein formation. The impairment of these two cellular defense systems against nitrosative stress resulted in different sets of S-nitrosated proteins, as revealed by the proteomics approach. CONCLUSION: Our results suggest that pharmacological intervention with mithramycin or auranofin may constitute promising tools for altering SNO homeostasis in tumour cells.


Subject(s)
Auranofin/pharmacology , Plicamycin/analogs & derivatives , Proteins/metabolism , S-Nitrosoglutathione/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Antirheumatic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Nitrosation , Plicamycin/pharmacology , Proteomics , Spectrometry, Mass, Electrospray Ionization , Tumor Cells, Cultured
20.
Lab Invest ; 90(1): 116-27, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19806079

ABSTRACT

Cholestatic liver injury following extra- or intrahepatic bile duct obstruction causes nonparenchymal cell proliferation and matrix deposition leading to end-stage liver disease and cirrhosis. In cholestatic conditions, nitric oxide (NO) is mainly produced by a hepatocyte-inducible NO synthase (iNOS) as a result of enhanced inflow of endotoxins to the liver and also by accumulation of bile salts in hepatocytes and subsequent hepatocellular injury. This study was aimed to investigate the role of NO and S-nitrosothiol (SNO) homeostasis in the development of hepatocellular injury during cholestasis induced by bile duct ligation (BDL) in rats. Male Wistar rats (200-250 g) were divided into four groups (n=10 each), including sham-operated (SO), bile duct-ligated (BDL), tauroursodeoxycholic acid (TUDCA, 50 mg/kg) and S-methylisothiourea (SMT, 25 mg/kg) treated. After 7 days, BDL rats showed elevated serum levels of gamma-glutamiltranspeptidase, aspartate aminotransferase, alanine aminotransferase, LDH, and bilirubin, bile duct proliferation and fibrosis, compared with the SO group. TUDCA treatment did not significantly alter these parameters, but the iNOS inhibitor SMT ameliorated hepatocellular injury, as shown by lower levels of circulating hepatic enzymes and bilirubin, and a decreased grade of bile duct proliferation and fibrosis. Both TUDCA and SMT treatments reversed Mrp2 canalicular pump expression to control levels. However, only SMT treatment significantly lowered the increased levels of plasma NO and S-nitrosation (S-nitrosylation) of liver proteins in BDL rats. Moreover, BDL resulted in a reduction of the S-nitrosoglutathione reductase (GSNOR/Adh5) enzymatic activity and a downregulation of the GSNOR/Adh5 mRNA expression that was reverted by SMT, but not TUDCA, treatment. A total of 25 liver proteins, including S-adenosyl methionine synthetase, betaine-homocysteine S-methyltransferase, Hsp90 and protein disulfide isomerase, were found to be S-nitrosated in BDL rats. In conclusion, the inhibition of NO production during induced cholestasis ameliorates hepatocellular injury. This effect is in part mediated by the improvement of cell proficiency in maintaining SNO homeostasis.


Subject(s)
Cholestasis/metabolism , Homeostasis/drug effects , Liver/metabolism , Liver/pathology , Nitric Oxide/antagonists & inhibitors , S-Nitrosothiols/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , Aldehyde Oxidoreductases/antagonists & inhibitors , Aldehyde Oxidoreductases/genetics , Animals , Bile Ducts , Cholagogues and Choleretics/pharmacology , Cholestasis/etiology , Cholestasis/pathology , Down-Regulation , Enzyme Inhibitors/pharmacology , Isothiuronium/analogs & derivatives , Isothiuronium/pharmacology , Ligation , Male , Nitric Oxide/biosynthesis , Nitric Oxide/blood , Nitrosation/drug effects , Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Taurochenodeoxycholic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...