Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Semin Cancer Biol ; 60: 262-273, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31654711

RESUMO

Brain, the major organ of the central nervous system controls and processes most of body activities. Therefore, the most aggressive brain tumor - glioblastoma and metastases from other organs to the brain are lethal leaving the patients with very short time of survival. The brain tissue landscape is very different from any other tissues and the specific microenvironment, comprising stem cells niches and blood-brain barrier, significantly influences the low rate of glioblastoma metastasis out of the brain, but better accommodates brain-invading cancer. In contrast to low frequency (0.5%) of all glioblastoma metastases, 10%-45% of other primary cancers do metastasize to the brain. This review addresses general cellular and molecular pathways that are to some extent similar in both types of metastases, involving circulating tumor cells (CTCs) with cancer stem cells (CSCs) characteristics, and metastatic niches. The invasion is a dynamic process involving reversible epithelial-to-mesenchymal (EMT) cell process, creating a transient gradient state that is inter-connected with epigenetic plasticity of the metastasizing (m)CSCs. These cells can switch between stationary, low proliferating/dormant state to a migratory, mesenchymal-like state. Settling in their respective niches as dormant CSCs in the secondary organ is a common feature in all types of metastases. In glioblastoma metastasis, the malignant mGSC cells express markers of mesenchymal GSC subtype (MES-GSC), such as CD44 and YK-40 and their major obstacle seems to be propagating in the in various organs' microenvironments, different from the niches that home GSCs in the primary glioblastoma. Focusing on one stromal component in the glioblastoma niches, the mesenchymal stem cells (MSCs), we report herein on their differential effects on glioblastoma cells, highly depending on their genetic subtype. On the other hand, in brain metastases, the major hindrance to metastatic progression of mCSCs seem to be crossing the blood-brain-barrier. Novel therapeutic approaches for brain metastases from various cancer types are advancing slowly, and the general trends involve targeting metastatic sub-clones and selective determinants of their niches. The update on the four most common brain metastases from lung, breast, melanoma and colorectal carcinoma is presented.


Assuntos
Neoplasias Encefálicas/secundário , Glioblastoma/etiologia , Animais , Biomarcadores , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Gerenciamento Clínico , Progressão da Doença , Suscetibilidade a Doenças , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Metástase Neoplásica , Células Neoplásicas Circulantes , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Nicho de Células-Tronco , Células Estromais/metabolismo , Células Estromais/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
2.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 594-603, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28040478

RESUMO

Glioblastoma (GBM) is the most aggressive primary brain tumor with poor patient survival that is at least partly caused by malignant and therapy-resistant glioma stem-like cells (GSLCs) that are protected in GSLC niches. Previously, we have shown that the chemo-attractant stromal-derived factor-1α (SDF-1α), its C-X-C receptor type 4 (CXCR4) and the cysteine protease cathepsin K (CatK) are localized in GSLC niches in glioblastoma. Here, we investigated whether SDF-1α is a niche factor that through its interactions with CXCR4 and/or its second receptor CXCR7 on GSLCs facilitates their homing to niches. Furthermore, we aimed to prove that SDF-1α cleavage by CatK inactivates SDF-1α and inhibits the invasion of GSLCs. We performed mass spectrometric analysis of cleavage products of SDF-1α after proteolysis by CatK. We demonstrated that CatK cleaves SDF-1α at 3 sites in the N-terminus, which is the region of SDF-1α that binds to its receptors. Confocal imaging of human GBM tissue sections confirmed co-localization of SDF-1α and CatK in GSLC niches. In accordance, 2D and 3D invasion experiments using CXCR4/CXCR7-expressing GSLCs and GBM cells showed that SDF-1α had chemotactic activity whereas CatK cleavage products of SDF-1α did not. Besides, CXCR4 inhibitor plerixafor inhibited invasion of CXCR4/CXCR7-expressing GSLCs. In conclusion, CatK can cleave and inactivate SDF-1α. This implies that CatK activity facilitates migration of GSLCs out of niches. We propose that activation of CatK may be a promising strategy to prevent homing of GSLCs in niches and thus render these cells sensitive to chemotherapy and radiation.


Assuntos
Catepsina K/metabolismo , Quimiocina CXCL12/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neuroglia/metabolismo , Receptores CXCR4/metabolismo , Sequência de Aminoácidos , Benzilaminas , Catepsina K/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/química , Quimiocina CXCL12/genética , Quimiotaxia , Ciclamos , Expressão Gênica , Compostos Heterocíclicos/farmacologia , Humanos , Células-Tronco Neoplásicas/patologia , Neuroglia/patologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteólise , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Nicho de Células-Tronco/genética
3.
Radiol Oncol ; 52(2): 152-159, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30018518

RESUMO

BACKGROUND: Detection of circulating lung cancer cells with cancer-stem like characteristics would represent an improved tool for disease prognosis. However, current antibodies based methods have some disadvantages and therefore cell SELEX (Systematic Evolution of Ligands by Exponential Enrichment) was used to develop DNA aptamers, recognizing cell surface markers of non-small lung carcinoma (NSLC) cells. MATERIALS AND METHODS: The human adenocarcinoma cell line A549 was used for selection in seven cell SELEX cycles. We used human blood leukocytes for negative selection, and lung stem cell protein marker CD90 antibody binding A549 cells for positive selection. RESULTS: The obtained oligonucleotide sequences after the seventh SELEX cycle were subjected to in silico selection analysis based on three independent types of bioinformatics approaches, selecting two closely related aptamer candidates in terms of consensus sequences, structural motifs, binding affinity (Kd) and stability (ΔG). We selected and identified the aptamer A155_18 with very good binding characteristics to A459 cells, selected for CD90 antibody binding. The calculated phylogenetic tree showed that aptamers A155_18 and the known A549 cell aptamer S6 have a close structural relationship. MEME sequence analysis showed that they share two unique motifs, not present in other sequences. CONCLUSIONS: The novel aptamer A155_18 has strong binding affinity for A549 lung carcinoma cell line subpopulation that is expressing stem cell marker CD90, indicating a possible stemness, characteristic for the A459 line, or a subpopulation present within this cell line. This aptamer can be applied as diagnostic tool, identifying NSLC circulating cells.

4.
Semin Cancer Biol ; 35: 71-84, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26320409

RESUMO

Proteases, including lysosomal cathepsins, are functionally involved in many processes in cancer progression from its initiation to invasion and metastatic spread. Only recently, cathepsin K (CatK), the cysteine protease originally reported as a collagenolytic protease produced by osteoclasts, appeared to be overexpressed as well in various types of cancers. In this review, the physiological functions of CatK are presented and compared to its potential role in pathobiolology of processes associated with tumour growth, invasion and metastasis of cancer cells and their interactions with the tumour microenvironment. CatK activity is either indirectly affecting signalling pathways, or directly degrading extracellular matrix (ECM) proteins, for example in bone metastases. Recently, CatK was also found in glioma, possibly regulating cancer stem-like cell mobilisation and modulating recently found physiological CatK substrates, including chemokines and growth factors. Moreover, CatK may be useful in differential diagnosis and may have prognostic value. Finally, the application of CatK inhibitors, which are already in clinical trials for treatment of osteoporosis, has a potential to attenuate cancer aggressiveness.


Assuntos
Catepsina K/metabolismo , Neoplasias/metabolismo , Peptídeo Hidrolases/metabolismo , Animais , Biomarcadores Tumorais , Catepsina K/antagonistas & inibidores , Catepsina K/genética , Progressão da Doença , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Peptídeo Hidrolases/genética , Prognóstico , Proteólise , Resultado do Tratamento , Microambiente Tumoral/genética
5.
Cytometry A ; 89(4): 365-75, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26671187

RESUMO

The most aggressive subtype of brain tumors is glioma WHO grade IV, the glioblastoma (GBM). The present work aims to elucidate the role of kinin receptors in interactions between GBM cells and mesenchymal stem cells (MSC). The GBM cell line U87-MG was stably transfected to express dsRed protein, single cell cloned, expanded, and cultured with MSC, both in the direct co-cultures (DC) and indirect co-cultures (IC) at equal cell number ratio for 72 h. Up- and down-regulation of matrix metalloproteases (MMP)-9 expression in U87-MG and MSC cells, respectively, in direct co-culture points to possible MSC participation in tumor invasion. MMP9 expression is in line with significantly increased expression of kinin B1 (B1R) and B2 receptor (B2R) in U87-MG cells and their decreased levels in MSC, as confirmed by quantitative assessment using flow cytometric analysis. Similarly, in indirect cultures (IC), lacking the contact between GBM and MSC cells, an increase of B1 and B2 receptor expression was again noted in U87-MG cells, and no significant changes in kinin receptors in MSC was observed. Functionality of kinin-B1 and B2 receptors was evidenced by stimulation of intracellular calcium fluxes by their respective agonists, des-Arg9-bradykinin (DBK) and bradykinin (BK). Moreover, BK showed a feedback control on kinin receptor expression in mono-cultures, direct and indirect co-cultures. The treatment with BK resulted in down-regulation of B1 and B2 receptors in MSC, with simultaneous up-regulation of these receptors in U87-MG cells, suggesting that functions of BK in information flow between these cells is important for tumor progression and invasion. © 2015 International Society for Advancement of Cytometry.


Assuntos
Bradicinina/metabolismo , Glioblastoma/metabolismo , Células-Tronco Mesenquimais/citologia , Receptores da Bradicinina/metabolismo , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Humanos , Transdução de Sinais/fisiologia , Regulação para Cima
6.
Radiol Oncol ; 50(2): 159-67, 2016 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27247548

RESUMO

BACKGROUND: An attractive approach in the study of human cancers is the use of transparent zebrafish (Danio rerio) embryos, which enable the visualization of cancer progression in a living animal. MATERIALS AND METHODS: We implanted mixtures of fluorescently labeled glioblastoma (GBM) cells and bonemarrow-derived mesenchymal stem cells (MSCs) into zebrafish embryos to study the cellular pathways of their invasion and the interactions between these cells in vivo. RESULTS: By developing and applying a carbocyanine-dye-compatible clearing protocol for observation of cells in deep tissues, we showed that U87 and U373 GBM cells rapidly aggregated into tumor masses in the ventricles and midbrain hemispheres of the zebrafish embryo brain, and invaded the central nervous system, often using the ventricular system and the central canal of the spinal cord. However, the GBM cells did not leave the central nervous system. With co-injection of differentially labeled cultured GBM cells and MSCs, the implanted cells formed mixed tumor masses in the brain. We observed tight associations between GBM cells and MSCs, and possible cell-fusion events. GBM cells and MSCs used similar invasion routes in the central nervous system. CONCLUSIONS: This simple model can be used to study the molecular pathways of cellular processes in GBM cell invasion, and their interactions with various types of stromal cells in double or triple cell co-cultures, to design anti-GBM cell therapies that use MSCs as vectors.

7.
Cytometry A ; 87(9): 806-16, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26189784

RESUMO

Aptamers are short single-stranded nucleic acids (RNA or ssDNA), identified by an in vitro selection process, denominated SELEX, from a partially random oligonucleotide library. They bind to a molecular target, a protein or other complex macromolecular structures of interest with high affinity and specificity, comparable to those of antibodies. Recently, aptamer selection protocols were developed for targeting living cells, including tumors. Chemical modifications of the aptamers and modalities of their detection and delivery systems are already available with high selectivity and targeting ability for the desired cancer cell type, making them promising for diagnosis and therapy. Glioblastoma multiformae represents the most malignant and fatal stage of glioma, and is also the most frequent brain tumor. Glioblastoma-specific aptamers were developed by either targeting the whole cell surface or known glioma biomarkers. These aptamers may gain importance for imaging, tumor cell isolation from biopsies and drug delivery. In biomedical imaging techniques, aptamers coupled with radionuclide or fluorescent labels, bioconjugates and nanoparticles offer an advanced, noninvasive manner for defining the glioblastoma tissue border. Though single modality aptamer imaging probes have some limitations, these are overcome by the use of multimodal probes. Due to selectivity and chemical characteristics, aptamers can be coupled to functionalized nanoparticles and loaded with a drug, appeared promising for in vivo targeting of glioblastoma. Finally, aptamers are effective mediators for gene silencing when coupled to small interfering RNA and a viral vector, thus providing a novel tool with enhanced targeting capability in drug delivery, designed for tailored treatment of glioblastoma patients.


Assuntos
Aptâmeros de Nucleotídeos/administração & dosagem , Aptâmeros de Nucleotídeos/genética , Neoplasias Encefálicas/genética , Diagnóstico por Imagem/métodos , Marcação de Genes/métodos , Glioblastoma/genética , Animais , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Glioblastoma/diagnóstico , Glioblastoma/terapia , Humanos , Técnica de Seleção de Aptâmeros/métodos
8.
Radiol Oncol ; 47(4): 330-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24294177

RESUMO

BACKGROUND: Patient-derived glioblastoma (GBM) stem-like cells (GSCs) represent a valuable model for basic and therapeutic research. GSCs are usually propagated in serum-free Neural Basal medium supplemented with bFGF and EGF. Yet, the exact influence of these growth factors on GSCs is still unclear. Recently it was suggested that GBM stem-like cells with amplified EGFR should be cultured in stem cell medium without EGF, as the presence of EGF induced rapid loss of EGFR amplification. However, patient biopsies are usually taken into culture before their genomic profiles are defined. Thus, an important question remains whether GBM cells without EGFR amplification also can be cultured in stem cell medium without EGF. MATERIALS AND METHODS: [corrected] To address this question, we used two heterogeneous glioblastoma GSC lines (NCH421k and NCH644) that lack EGFR amplification. RESULTS: Although both cell lines showed very low EGFR expression under standard growth conditions, bFGF stimulation induced higher expression of EGFR in NCH644. In both cell lines, expression of the stem cell markers nestin and CD133 was higher upon stimulation with bFGF compared to EGF. Importantly, bFGF stimulated the growth of both cell lines, whereas EGF had no effect. We verified that the growth stimulation by bFGF was either mediated by proliferation (NCH421k) or resistance to apoptosis (NCH644). CONCLUSIONS: We demonstrate that GSC cultures without EGFR amplification can be maintained and expanded with bFGF, while the addition of EGF has no significant effect and therefore can be omitted.

9.
Int J Cancer ; 131(8): 1779-89, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22287159

RESUMO

Cysteine cathepsins play an important role in shaping the highly infiltrative growth pattern of human gliomas. We have previously demonstrated that the activity of cysteine cathepsins is elevated in invasive glioblastoma (GBM) cells in vitro, in part due to attenuation of their endogenous inhibitors, the cystatins. To investigate this relationship in vivo, we established U87-MG xenografts in non-obese diabetic (NOD)/severe combined immunodeficiency (SCID)-enhanced green fluorescent protein (eGFP) mice. Here, tumor growth correlated with an elevated enzymatic activity of CatB both in the tumor core and at the periphery, whereas CatS and CatL levels were higher at the xenograft edge compared to the core. Reversely, StefB expression was detected in the tumor core, but it was generally absent in the tumor periphery, suggesting that down-regulation of this inhibitor correlates with in vivo invasion. In human GBM samples, all cathepsins were elevated at the tumor periphery compared to brain parenchyma. CatB was also typically associated with angiogenic endothelia and necrotic areas. StefB was mainly detected in the tumor core, whereas CysC and StefA were evenly distributed, reflecting the observations in the xenografts. However, at the mRNA level, no differences in cathepsins and cystatins were observed between the tumor center and the periphery in both human biopsies and xenografts. Interestingly, in human tumors, cathepsin and stefin transcript levels correlated with CD68 and CXCR4 levels, but not with epidermal growth factor receptor (EGFR). Moreover, we reveal for the first time that an elevated StefA mRNA level is a highly significant prognostic factor for patient survival.


Assuntos
Biomarcadores Tumorais/metabolismo , Catepsinas/metabolismo , Cistatinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/patologia , Animais , Biomarcadores Tumorais/genética , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Catepsinas/genética , Cistatinas/genética , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Glioblastoma/genética , Glioblastoma/mortalidade , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida
10.
Front Oncol ; 12: 965882, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36119523

RESUMO

Venoms are complex mixtures of different molecules and ions. Among them, bioactive peptides have been found to affect cancer hallmarks, such as cell proliferation, cell invasion, cell migration, and can also modulate the immune response of normal and cancer-bearing organisms. In this article, we review the mechanisms of action on these cancer cell features, focusing on bioactive peptides being developed as potential therapeutics for one of the most aggressive and deadly brain tumors, glioblastoma (GB). Novel therapeutic approaches applying bioactive peptides may contribute to multiple targeting of GB and particularly of GB stem cells. Bioactive peptides selectively target cancer cells without harming normal cells. Various molecular targets related to the effects of bioactive peptides on GB have been proposed, including ion channels, integrins, membrane phospholipids and even immunomodulatory treatment of GB. In addition to therapy, some bioactive peptides, such as disintegrins, can also be used for diagnostics or are used as labels for cytotoxic drugs to specifically target cancer cells. Given the limitations described in the last section, successful application in cancer therapy is rather low, as only 3.4% of such peptides have been included in clinical trials and have passed successfully phases I to III. Combined approaches of added bioactive peptides to standard cancer therapies need to be explored using advanced GB in vitro models such as organoids. On the other hand, new methods are also being developed to improve translation from research to practice and provide new hope for GB patients and their families.

11.
Cancers (Basel) ; 14(23)2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36497400

RESUMO

Glioblastoma (GBM) is one of the most aggressive cancers, comprising 60-70% of all gliomas. The large G-protein-coupled receptor family includes cannabinoid receptors CB1, CB2, GPR55, and non-specific ion receptor protein transporters TRPs. First, we found up-regulated CNR1, GPR55, and TRPV1 expression in glioma patient-derived tissue samples and cell lines compared with non-malignant brain samples. CNR1 and GPR55 did not correlate with glioma grade, whereas TRPV1 negatively correlated with grade and positively correlated with longer overall survival. This suggests a tumour-suppressor role of TRPV1. With respect to markers of GBM stem cells, preferred targets of therapy, TRPV1 and GPR55, but not CNR1, strongly correlated with different sets of stemness gene markers: NOTCH, OLIG2, CD9, TRIM28, and TUFM and CD15, SOX2, OCT4, and ID1, respectively. This is in line with the higher expression of TRPV1 and GPR55 genes in GSCs compared with differentiated GBM cells. Second, in a panel of patient-derived GSCs, we found that CBG and CBD exhibited the highest cytotoxicity at a molar ratio of 3:1. We suggest that this mixture should be tested in experimental animals and clinical studies, in which currently used Δ9-tetrahydrocannabinol (THC) is replaced with efficient and non-psychoactive CBG in adjuvant standard-of-care therapy.

12.
Cells ; 10(2)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562819

RESUMO

Glioblastoma is the most aggressive cancer among primary brain tumours. As with other cancers, the incidence of glioblastoma is increasing; despite modern therapies, the overall mean survival of patients post-diagnosis averages around 16 months, a figure that has not changed in many years. Cannabigerol (CBG) has only recently been reported to prevent the progression of certain carcinomas and has not yet been studied in glioblastoma. Here, we have compared the cytotoxic, apoptotic, and anti-invasive effects of the purified natural cannabinoid CBG together with CBD and THC on established differentiated glioblastoma tumour cells and glioblastoma stem cells. CBG and THC reduced the viability of both types of cells to a similar extent, whereas combining CBD with CBG was more efficient than with THC. CBD and CBG, both alone and in combination, induced caspase-dependent cell apoptosis, and there was no additive THC effect. Of note, CBG inhibited glioblastoma invasion in a similar manner to CBD and the chemotherapeutic temozolomide. We have demonstrated that THC has little added value in combined-cannabinoid glioblastoma treatment, suggesting that this psychotropic cannabinoid should be replaced with CBG in future clinical studies of glioblastoma therapy.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Canabinoides/uso terapêutico , Glioblastoma/tratamento farmacológico , Apoptose , Neoplasias Encefálicas/patologia , Canabinoides/farmacologia , Feminino , Glioblastoma/patologia , Humanos
13.
Biol Chem ; 391(5): 519-31, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20302512

RESUMO

Arsenic trioxide (arsenite) was the first chemotherapeutic drug to be described and is now being rediscovered in cancer treatment, including glioblastoma multiforme. Arsenite toxicity triggers autophagy in cancer cells, although final stages of the process involve executive caspases, suggesting an interplay between autophagic and apoptotic pathways that awaits to be explained at a molecular level. We evaluated the contribution of the lysosomal cathepsins (Cat) L and B, which are upregulated in glioblastomas, in the mechanism of arsenite toxicity in human glioblastoma cells. Arsenite treatment induced autophagosome formation and permeabilization of mitochondria, followed by caspase 3/7-mediated apoptosis. The autophagy inhibitor 3-methyladenine protected from arsenite toxicity, whereas bafilomycin A1 did not. Furthermore, arsenite significantly decreased CatB levels and selectively inhibited its cellular and recombinant protein activity, while not affecting CatL. However, downregulation of CatL greatly enhanced apoptosis by arsenite. Our results show that arsenite toxicity involves a complex interplay between autophagy and apoptosis in human glioblastoma cells and is associated with inhibition of CatB, and that this toxicity is highly exacerbated by simultaneous CatL inhibition. The latter points to a synergy that could be used in clinical treatment to lower the therapeutic dose, thus avoiding the toxic side effects of arsenite in glioblastoma management.


Assuntos
Antineoplásicos/uso terapêutico , Arsenicais/uso terapêutico , Autofagia , Catepsina B/fisiologia , Catepsina L/fisiologia , Glioblastoma/tratamento farmacológico , Óxidos/uso terapêutico , Adenina/análogos & derivados , Adenina/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais/efeitos adversos , Autofagia/efeitos dos fármacos , Caspases/metabolismo , Catepsina B/antagonistas & inibidores , Catepsina L/antagonistas & inibidores , Linhagem Celular Tumoral , Dipeptídeos/farmacologia , Regulação para Baixo , Compostos de Epóxi/farmacologia , Glioblastoma/metabolismo , Humanos , Macrolídeos/farmacologia , Óxidos/efeitos adversos , Piridinas/farmacologia
14.
Radiol Oncol ; 44(1): 42-51, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22933890

RESUMO

BACKGROUND: Human exposure to genotoxic agents in the environment and everyday life represents a serious health threat. Fast and reliable assessment of genotoxicity of chemicals is of main importance in the fields of new chemicals and drug development as well as in environmental monitoring. The tumor suppressor gene p21, the major downstream target gene of activated p53 which is responsible for cell cycle arrest following DNA damage, has been shown to be specifically up-regulated by genotoxic carcinogens. The aim of our study was to develop a human cell-based biosensor system for simple and fast detection of genotoxic agents. METHODS: Metabolically active HepG2 human hepatoma cells were transfected with plasmid encoding Enhanced Green Fluorescent Protein (EGFP) under the control of the p21 promoter (p21HepG2GFP). DNA damage was induced by genotoxic agents with known mechanisms of action. The increase in fluorescence intensity, due to p21 mediated EGFP expression, was measured with a fluorescence microplate reader. The viability of treated cells was determined by the colorimetric MTS assay. RESULTS: The directly acting alkylating agent methylmethane sulphonate (MMS) showed significant increase in EGFP production after 48 h at 20 µg/mL. The indirectly acting carcinogen benzo(a)pyren (BaP) and the cross-linking agent cisplatin (CisPt) induced a dose- dependent increase in EGFP fluorescence, which was already significant at concentrations 0.13 µg/mL and 0.41 µg/mL, respectively. Vinblastine (VLB), a spindle poison that does not induce direct DNA damage, induced only a small increase in EGFP fluorescence intensity after 24 h at the lowest concentration (0.1 µg/mL), while exposure to higher concentrations was associated with significantly reduced cell viability. CONCLUSIONS: The results of our study demonstrated that this novel assay based on the stably transformed cell line p21HepG2GFP can be used as a fast and simple biosensor system for detection of genetic damage caused by chemical agents.

15.
Biochim Biophys Acta Mol Cell Res ; 1867(10): 118782, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32554164

RESUMO

Epithelial-to-mesenchymal transition (EMT) is an essential molecular and cellular process that is part of normal embryogenesis and wound healing, and also has a ubiquitous role in various types of carcinoma and glioblastoma. EMT is activated and regulated by specific microenvironmental endogenous triggers and a complex network of signalling pathways. These mostly include epigenetic events that affect protein translation-controlling factors and proteases, altogether orchestrated by the switching on and off of oncogenes and tumour-suppressor genes in cancer cells. The hallmark of cancer-linked EMT is that the process is incomplete, as it is opposed by the reverse process of mesenchymal-to-epithelial transition, which results in a hybrid epithelial/mesenchymal phenotype that shows notable cell plasticity. This is a characteristic of cancer stem cells (CSCs), and it is of the utmost importance in their niche microenvironment, where it governs CSC migratory and invasive properties, thereby creating metastatic CSCs. These cells have high resistance to therapeutic treatments, in particular in glioblastoma.


Assuntos
Transição Epitelial-Mesenquimal , Glioblastoma/patologia , Microambiente Tumoral , Plasticidade Celular , Glioblastoma/diagnóstico , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia
16.
Radiol Oncol ; 53(4): 397-406, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31747383

RESUMO

Background Glioblastoma is the most frequent and aggressive brain tumour in humans with median survival from 12 to 15 months after the diagnosis. This is mostly due to therapy resistant glioblastoma stem cells in addition to intertumour heterogeneity that is due to infiltration of a plethora of host cells. Besides endothelial cells, mesenchymal stem cells and their differentiated progenies, immune cells of various differentiation states, including monocytes, comprise resident, brain tumour microenvironment. There are compelling evidence for CCL5/CCR5 in the invasive and metastatic behaviour of many cancer types. CCR5, a G-protein coupled receptor, known to function as an essential co-receptor for HIV entry, is now known to participate in driving tumour heterogeneity, the formation of cancer stem cells and the promotion of cancer invasion and metastasis. Clinical trials have recently opened targeting CCR5 using a humanized monoclonal antibody (leronlimab) for metastatic triple negative breast cancer (TNBC) or a small molecule inhibitor (maraviroc) for metastatic colon cancer. There are important CCL5 and CCR5 structure and signalling mechanisms in glioblastoma. In addition, the CCL5/CCR5 axis directs infiltration and interactions with monocytes/macrophages and mesenchymal stem cells, comprising glioblastoma stem cell niches. Conclusions CCR5 is highly expressed in glioblastoma and is associated with poor prognosis of patients. CCL5/CCR5 is suggested to be an excellent new target for glioblastoma therapy. The molecular mechanisms, by which chemoattractant and receptor respond within the complex tissue microenvironment to promote cancer stem cells and tumour heterogeneity, should be considered in forthcoming studies.


Assuntos
Neoplasias Encefálicas/imunologia , Antagonistas dos Receptores CCR5/uso terapêutico , Quimiocina CCL5/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Glioblastoma/imunologia , Receptores CCR5/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Antagonistas dos Receptores CCR5/farmacologia , Progressão da Doença , Glioblastoma/patologia , Humanos , Terapia de Alvo Molecular , Metástase Neoplásica , Microambiente Tumoral
17.
Cancer Microenviron ; 12(2-3): 77-94, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31420805

RESUMO

Tumour progression involves interactions among various cancer cell clones, including the cancer stem cell subpopulation and exogenous cellular components, termed cancer stromal cells. The latter include a plethora of tumour infiltrating immunocompetent cells, among which are also immuno-modulatory mesenchymal stem cells, which by vigorous migration to growing tumours and susequent transdifferentiation into various types of tumour-residing stromal cells, may either inhibit or support tumour progression. In the light of the scarce therapeutic options existing for the most malignant brain tumour glioblastoma, mesenchymal stem cells may represent a promising novel tool for cell therapy, e.g. drug delivery vectors. Here, we review the increasing number of reports on mutual interactions between mesenchymal stem cells and glioblastoma cells in their microenvironment. We particularly point out two novel aspects: the different responses of cancer cells to their microenvironmental cues, and to the signalling by kinin receptors that complement the immuno-modulating cytokine-signalling networks. Inflammatory glioblastoma microenvironment is characterised by increasing expression of kinin receptors during progressive glioma malignancy, thus making kinin signalling and kinins themselves rather important in this context. In general, their role in tumour microenvironment has not been explored so far. In addition, kinins also regulate blood brain barrier-related drug transfer as well as brain tumour angiogenesis. These studies support the on-going research on kinin antagonists as candidates in the development of anti-invasive agents for adjuvant glioblastoma therapy.

18.
Toxicon ; 51(4): 615-23, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18191168

RESUMO

Microcystins (MCs) are hepatotoxic cyclic heptapeptides produced by freshwater cyanobacteria. They are inhibitors of serine/threonine protein phosphatases 1A and 2A and are involved in liver tumour promotion. Several recent studies indicated that MCs are genotoxic and may also act as tumour initiators. Based on our previous results showing that microcystin-LR (MCLR) induces DNA damage in HepG2 cells, we have now explored the effect of MCLR on the expression of selected genes known to be involved in the cell response to DNA damage and apoptosis. The HepG2 cells were exposed to non-cytotoxic concentrations (0.01, 0.1 and 1 microg/ml) of MCLR for various periods of time (2-16 h) and the mRNA expression was determined with the quantitative real-time polymerase chain reaction (QRT-PCR). We found a significantly elevated expression of tumour suppressor gene p53 and its downstream-regulated genes involved in DNA repair and cell cycle regulation (p21, gadd 45a, mdm2), as well as increased expression of the pro-apoptotic gene bax, but no alterations of the anti-apoptotic bcl-2. Up-regulation of the expression of mdm2, p21 and gadd45a provides strong support for our previous suggestion that MCLR is a genotoxic carcinogen. The increased ratio of expression of bax to that of bcl-2 induced by MCLR suggests that apoptosis in HepG2 cells proceeds via the mitochondrial pathway.


Assuntos
Apoptose/fisiologia , Dano ao DNA/fisiologia , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Microcistinas/farmacologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Epiteliais/fisiologia , Humanos , Neoplasias Hepáticas , Toxinas Marinhas , Fatores de Tempo
19.
Toxicon ; 52(3): 518-25, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18657565

RESUMO

Microcystins, which are hepatotoxins produced by cyanobacteria, have been reported to be potent tumour promoters, and there is an indication that they can also act as tumour initiators. They thus constitute a potential threat to human and animal health, at concentrations that do not cause acute hepatotoxic effects. The main target organ of microcystin toxicity is the liver; however, several studies have shown that other organs and tissues may also be affected. We have investigated the effect of non-cytotoxic concentrations of microcystin-LR (MCLR) on the generation of intracellular reactive oxygen species (ROS) and on DNA damage in human colon adenocarcinoma CaCo-2, human astrocytoma IPDDC-A2 and human B-lymphoblastoid NCNC cell lines. The viability of CaCo-2 cells exposed to 10 microg/MCLR for 24 and 48 h was reduced by about 40%, while that of NCNC and IPDDC-2A cells was not affected. Intracellular ROS production was increased in CaCo-2 and IPDDC-2A, but not NCNC, cells. Using the comet assay, it was shown that MCLR, at non-cytotoxic concentrations, induced a time and dose dependent increase of DNA damage in CaCo-2 cells, but not significantly in IPDDC-2A and NCNC cells. Thus, CaCo-2 cells were the most sensitive. Their sensitivity is comparable to that observed in our previous study with human hepatoma HepG2 cells. These results indicate that, in addition to liver cells, colon cells should also be considered as a target for microcystin toxicity, and that exposure to low doses of microcystins may affect intestinal tissue.


Assuntos
Dano ao DNA/efeitos dos fármacos , Microcistinas/farmacologia , Adenocarcinoma , Astrocitoma , Linfócitos B , Linhagem Celular Tumoral , Humanos , Toxinas Marinhas , Espécies Reativas de Oxigênio
20.
Sci Rep ; 8(1): 1299, 2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29358738

RESUMO

Glioblastoma multiforme (GBM) represents the most lethal brain tumour, and these tumours have very limited treatment options. Mesenchymal stem cells (MSC) are considered as candidates for advanced cell therapies, due to their tropism towards GBM, possibly affecting their malignancy, thus also representing a potential therapeutic vector. Therefore, we aimed to compare the effects of bone-marrow-derived versus adipose-tissue-derived MSC (BM-/AT-MSC) on heterogeneous populations of tumour cells. This cells' interplay was addressed by the in-vitro two-dimensional (monolayer) and three-dimensional (spheroid) co-culture models, using U87 and U373 GBM cell lines, expressing genotypically different mesenchymal transcriptome profiles. U87 cell low mesenchymal profile expressed high levels of kinin receptor 1 (B1R) and their invasion was greatly enhanced by the B1R agonist des-Arg9-bradykinin upon BM-MSC co-culturing in 3D co-cultures. This correlated to significantly higher cell-cell interactions in U87/BM-MSC mixed spheroids. This was not observed with the U373 cells and not in AT-MSC co-cultures. Altogether, these data support the on-going exploration of B1R as target for adjuvant approach in GBM therapy. Secondly, the results emphasize the need for further careful exploration of the selectivity regarding the origin of MSC as potential candidates for cell therapies, particular in cancer, where they may adversely affect heterogeneous tumour cell populations.


Assuntos
Bradicinina/análogos & derivados , Comunicação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Receptor B1 da Bradicinina/agonistas , Esferoides Celulares/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Bradicinina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Especificidade de Órgãos , Receptor B1 da Bradicinina/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/efeitos dos fármacos , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa