Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biofabrication ; 16(2)2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38377605

RESUMO

The current cancer research and drug testing are primarily based on 2D cell cultures and animal models. However, these methods have limitations and yield distinct drug response patterns. This study addressed this gap by developing an innovativein vitrohuman three-dimensional (3D) normal skin model and a multicellular model of human cutaneous squamous cell carcinoma (cSCC) using 3D bioprinting technology. Comparative analyzes were performed between bioprinted 3D-cSCC model, consisting of HaCaT keratinocytes, primary normal human dermal fibroblasts and A431 cancer cells (tricellular), bioprinted 3D-A431 model composed of A431 cancer cells only (monocellular), A431 cancer cell spheroids, and conventional 2D models. The models were structurally characterized by light microscopy, immunofluorescence (LIVE/DEAD assay, confocal microscopy) and immunohistochemistry (hematoxylin/eosin, p63, vimentin, Ki67, epidermal growth factor receptor stainings). The spatial arrangement of the 3D models was analyzed using the ARIVIS scientific image analysis platform. All models were also functionally assessed by cetuximab (CTX) response testing with the MTS assay. 3D-cSCC models were maintained for up to 16 weeks. Morphological and histological examinations confirmed the presence of skin-like layers in the bioprinted 3D models of normal skin, and the intricate and diverse features of the bioprinted skin cancer model, replicating the criticalin vivocharacteristics. In both mono- and tricellular bioprinted tumor constructs, there was a gradual formation and continuous growth of spheroid-like clusters of cancer cells, significantly influencing the morphology of the models. Cancer cells in the 3D bioprinted constructs showed reduced sensitivity to CTX compared to spheroids and 2D cultures. This study underscores the potential of 3D multicellular models in elucidating drug responses and gaining a better understanding the intricate interplay of cellular components within the tumor microenvironment. Developing the multicellular 3D tumor model paves the way for new research critical to advancing fundamental cancer research and future clinical applications, particularly drug response testing.


Assuntos
Bioimpressão , Carcinoma de Células Escamosas , Neoplasias Cutâneas , Animais , Humanos , Técnicas de Cultura de Células/métodos , Pele , Queratinócitos , Bioimpressão/métodos , Esferoides Celulares , Impressão Tridimensional , Microambiente Tumoral
2.
J Steroid Biochem Mol Biol ; 189: 48-62, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30772447

RESUMO

Conventional cytotoxic drugs preferentially eliminate differentiated cancer cells but spare relatively more resistant stem-like cancer cells capable to initiate recurrence. Due to cancer cell plasticity, the stem-like phenotype can be also acquired by cancer cells refractory to treatment with cytotoxic drugs. We investigated whether drugs inhibiting receptor tyrosine kinases could be used to target human colon cancer cells initiating cancer regrowth following conventional cytotoxic treatment. The moderately differentiated cell line HT-29 and poorly differentiated cell line HCT-116 were exposed to 5-fluorouracil (5-FU). Cells that resisted the exposure to 5-FU were subsequently treated with imatinib or sunitinib. Both drugs reduced clonogenicity of 5-FU-refractory cells under normoxic and hypoxic culture conditions. The expression of numerous stemness-related genes was upregulated in cancer cells following the exposure to 5-FU, and remained at a high level in 5-FU-refractory cells undergoing renewal under normoxia, but decreased spontaneously under hypoxia. Imatinib downregulated the expression of stemness-related genes in cells undergoing renewal under normoxia. A combination of imatinib with PRI-2191, an analogue of 1,25-dihydroxyvitamin D3, downregulated stemness-related genes in HCT-116/5-FU cells more efficiently than imatinib alone. A synthetic analogue of 1,25-dihydroxyvitamin D2 (PRI-1906) abolished the effect of imatinib on gene expression in HCT-116/5-FU cells undergoing renewal under normoxia. Sunitinib promoted shift of phenotype of HT-29/5-FU cells undergoing renewal toward stem-like one. It suggests that the phenotype shift toward stemness induced by sequential sunitinib treatment following 5-FU treatment could increase a risk of cancer recurrence. In contrast to sunitinib, imatinib could be used both to interfere with cancer regrowth after conventional chemotherapy and to downregulate the expression of stemness-related genes in residual colon cancer cells capable to initiate cancer recurrence. The findings suggest that imatinib could also be combined with vitamin D analogue PRI-2191 to prevent recurrence more efficiently than imatinib alone and to compensate for vitamin D deficiency resulting from imatinib treatment.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Di-Hidroxicolecalciferóis/farmacologia , Fluoruracila/farmacologia , Mesilato de Imatinib/farmacologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HT29 , Humanos
3.
Arch Immunol Ther Exp (Warsz) ; 65(1): 51-67, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26956644

RESUMO

Conventional anti-cancer drugs preferentially eliminate differentiated cancer cells but those cells that are spared (i.e. cancer stem cells: CSC), initiate recurrence. We tested whether drugs that target receptor tyrosine kinases (RTKs) involved in developmental signaling cascades and activated in CSC, could be used to silence and/or to eliminate colorectal cancer cells refractory to conventional treatment with cytoreductive drugs. A sequential treatment model was thereby developed with doxorubicin (DOX) and imatinib. CT-26 mouse colon carcinoma cells were pre-treated with DOX to select DOX-refractory cells with CSC properties, which were then subsequently treated with RTK inhibitor imatinib, where their regrowth was found to be inhibited. Under both normoxic and hypoxic conditions, imatinib potently inhibited clonogenicity of DOX-refractory CT-26 cells. Treatment with DOX did not eliminate tumorigenic CT-26 cells, since CT-26 cells pre-exposed to DOX in vitro, when inoculated subcutaneously, induced tumors in 90 % of mice, as opposed to a 100 % rate in the case of chemonaive CT-26 cells. In mice inoculated with chemonaive CT-26 cells, tumor formation was not prevented by imatinib. However, imatinib prevented tumor formation in 50 % of mice inoculated with CT-26 cells pre-exposed to DOX in vitro, with the remaining 50 % mice showing delayed tumor formation. These results suggest that the sequential use of the drug imatinib, as a drug targeting cancer cells expressing stem cell features after conventional cytoreductive treatment, is a promising future strategy for preventing tumor recurrence.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Doxorrubicina/uso terapêutico , Mesilato de Imatinib/uso terapêutico , Antígeno AC133/química , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Citometria de Fluxo , Hipóxia , Camundongos , Camundongos Endogâmicos BALB C , Recidiva Local de Neoplasia , Células-Tronco Neoplásicas/efeitos dos fármacos , Oxigênio/química , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
4.
Int J Mol Sci ; 17(6)2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27314328

RESUMO

This study aimed to evaluate the capacity of hypocalcemic analogues of 1α,25-dihydroxyvitamin D2 (1,25D2) and 1α,25-dihydroxyvitamin D3 (1,25D3) to inhibit regrowth and regulate the stemness-related gene expression in colon cancer cells undergoing renewal after exposure to 5-fluorouracil (5-FU). All of the tested analogues of 1,25D2 equally potently decreased the clonogenicity and the proliferative activity of HT-29 cells which survived the exposure to 5-FU, but differently regulated gene expression of these cells during their renewal. 1,25D2 and analogues (PRI-1907 and PRI-1917), as well as 1,25D3 and analogue PRI-2191, decreased the relative expression level of several stemness-related genes, such as NANOG, OCT3/4, PROM1, SOX2, ALDHA1, CXCR4, in HT-29/5-FU cells during their renewal, in comparison to untreated HT-29/5-FU cells. The other 1,25D2 analogues (PRI-1906 and PRI-1916) were not capable of downregulating the expression of these stemness-related genes as the analogues PRI-1907 and PRI-1917 did. All of the tested vitamin D analogues upregulated CDH1, the gene encoding E-cadherin associated with epithelial phenotype. Out of the series of analogues studied, side-chain branched analogues of 1,25D2 (PRI-1907, PRI-1917) and the analogue of 1,25D3 (PRI-2191) might be used to target cancer cells with stem-like phenotypes that survive conventional chemotherapy.


Assuntos
Calcitriol/farmacologia , Vitaminas/farmacologia , Antineoplásicos/toxicidade , Calcitriol/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/toxicidade , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos
5.
Tumour Biol ; 37(4): 4699-709, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26511971

RESUMO

This study was aimed to determine whether hypocalcemic analogs of active forms of vitamins D modulate expression of genes related to stem-like phenotype in colon cancer cell lines HT-29 and HCT-116 undergoing renewal after the treatment with 5-fluorouracil (5-FU). Both lines express vitamin D receptor, but differ in differentiation stage and vitamin D sensitivity. Cells that resisted the 5-FU exposure were treated with synthetic analog of 1,25-dihydroxyvitamin D2 (PRI-1906) and analogs of 1,25-dihydroxyvitamin D3 (PRI-2191 and PRI-2205). Proliferative activity was more profoundly affected by vitamin D analogs in HT-29/5-FU than in HCT-116/5-FU cells. In HT-29/5-FU cells, analogs PRI-1906 and PRI-2191 downregulated the expression of genes related to survival, re-growth, and invasiveness during renewal, while PRI-2205 increased expression of genes related to differentiation only. In HCT-116/5-FU cells, PRI-2191 decreased the expression of stemness- and angiogenesis-related genes, whereas PRI-1906 augmented their expression. The effects in HCT-116/5-FU cells were observed at higher concentrations of the analogs than those used for HT-29/5-FU cells. Out of the series of analogs studied, PRI-2191 might be used to counteract the renewal of both moderately and poorly differentiated cancer cells following conventional treatment.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Calcitriol/análogos & derivados , Di-Hidroxicolecalciferóis/farmacologia , Resistencia a Medicamentos Antineoplásicos , Ergocalciferóis/farmacologia , Fluoruracila/farmacologia , Calcitriol/farmacologia , Autorrenovação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Sinergismo Farmacológico , Transição Epitelial-Mesenquimal , Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HT29 , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/fisiologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
6.
Arch Immunol Ther Exp (Warsz) ; 59(4): 315-26, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21633916

RESUMO

Post-radiation inflammatory reaction leads to an irreversible pulmonary fibrosis which may cause lethal respiratory insufficiency. Pathological inflammatory and fibrotic changes might be attenuated by inhibiting tumour necrosis factor (TNF)-α activity using TNF-α soluble receptors. Thus, an experimental antifibrotic gene therapy with the plasmid vector encoding a mouse soluble receptor I for TNF-α (psTNFR-I) was assessed. Soluble TNFR-I encoding gene was cloned into pcDNA3.1 plasmid. The ability of psTNFR-I expressing vector to transfect cells, and its biological activity in vitro and in vivo were examined by PCR, RT-PCR, MTT assay and ELISA. The C57Bl/6J mice received single intramuscular injection of psTNFR-I, conjugated with polyetylenimine (PEI) 25 kDa, equally divided to both hind legs, 3 days before irradiation (20 Gy, Co60), and either a single injection or ten injections once a week after irradiation. The data proved the effectiveness of psTNFR-I product to neutralise TNF-α activity in vitro. The in vivo plasmid incorporation and maintenance was confirmed. Measurements of plasma soluble TNFR-I levels showed that the in vivo gene transfer was effective. PEI was found to enhance transfection efficiency in vivo. The psTNFR-I/PEI complexes caused no toxicity in the transfected mice. C57Bl/6J mice that received prolonged psTNFR-I/PEI injections developed lethal fibrotic syndrome and died 8 weeks later than the mice treated with a double plasmid injection and the control mice treated with a control plasmid. Sequential administration of soluble TNFR-I by a nonviral, intramuscular gene transduction in the early and late post-radiation inflammatory phase prolonged survival of irradiated mice and attenuated the symptoms of lung fibrosis. The psTNFR-I gene transduction may provide a safe and simple method to partially neutralise TNF-α activity and prevent radiation-induced lung injury.


Assuntos
Pneumonite por Radiação/terapia , Receptores Tipo I de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Colágeno/análise , Modelos Animais de Doenças , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos , Receptores Tipo I de Fatores de Necrose Tumoral/administração & dosagem , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
Postepy Biochem ; 55(2): 121-8, 2009.
Artigo em Polonês | MEDLINE | ID: mdl-19824467

RESUMO

According to recently published data, the epithelial-mesenchymal transition--a process important for embryonic development, may be involved in many pathological processes such as wound healing, tissue fibrosis or cancer progression. The EMT process in cell is driven by growth factors (EGF, PDGF, HGF) or other signaling proteins such as TGF-beta, sonic hedgehog (Shh), Wnt/beta-catenin and extracellular matrix (ECM) components that may stimulate cellular growth and migration. During cancer progression, the EMT process is necessary to the conversion of benign tumor to aggressive and highly invasive cancer. This is due to complex changes in cancer cells and their microenvironment that lead to dissolution of intracellular junctions and their detachment from basolateral membrane, and changes in the interactions between cancer cells and ECM. The loss of adhesion is accompanied by molecular and morphologic changes in cancer cells that are essential for the phenotypic change from epithelial to mesenchymal one, and the acquirement of higher migration and invasion potential. During the colonization of distant sites, a reverse process mesenchymal-epithelial transition (MET) takes place and metastatic cancer cells again acquire the epithelial phenotype. The EMT in cancer progression is not only specific for cancer cells. It has been suggested that also cells within tumor microenvironment e.g. cancer associated fibroblasts (CAF) are generated in part from normal epithelial cells in EMT process. The understanding of the role of EMT and MET processes in cancer progression and their relationship with cancer stem cells, cancer associated fibroblasts and other stroma cells might lead to the discovery of new, targeted cancer therapies.


Assuntos
Transformação Celular Neoplásica/patologia , Células Epiteliais/patologia , Mesoderma/patologia , Neoplasias/patologia , Neoplasias/fisiopatologia , Animais , Anoikis , Progressão da Doença , Humanos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia
8.
Biochem Pharmacol ; 69(1): 73-85, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15588716

RESUMO

The aim of the present study was to investigate changes in spectrin and protein kinase C theta; (PKC theta;) organisation in human lymphoid and leukaemic cells undergoing chemotherapeutically induced apoptosis. An analysis of spectrin arrangement in human peripheral lymphoid (non-Hodgkin lymphoma) and leukaemic (acute lymphoblastic leukaemia) cells before and after chemotherapy revealed radical differences in the distribution of this protein. By using immunofluorescent technique, in lymphocytes isolated before chemotherapy, we found spectrin evenly distributed in the cytoplasm and the plasma membrane, while after the therapy changes in spectrin organisation occurred. Moreover, in lymphocytes after chemotherapy, extraction with buffer containing non-ionic detergent (Triton X-100) revealed presence of an insoluble fraction of spectrin. In normal or malignant cells before chemotherapy spectrin was totally soluble, however it should be mentioned that in total cell extracts and supernatants (but not in pellets) apoptotic fragments of spectrin (in addition to intact alpha and beta chains) were also found. In malignant cells after chemotherapy changes in PKC theta; organisation, similar to this observed in the case of spectrin, were shown by the immunofluorescence technique. In contrast, no differences in the distribution of other isoforms of protein kinase C: betaI and betaII, before and after chemotherapy, were found. Apoptotic phosphatidyloserine (PS) externalisation, as well as cell shrinkage, membrane protrusions and blebbing were observed in lymphocytes after chemotherapy and treatment with cytostatics in vitro. The overall results may suggest that spectrin redistribution/aggregation is the phenomenon involved in programmed cell death (PCD) of normal and neoplastic lymphocytes and lymphoblasts, however molecular basis of this phenomenon should be further investigated.


Assuntos
Linfócitos/metabolismo , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Espectrina/metabolismo , Células Cultivadas , Humanos , Linfócitos/química , Linfócitos/efeitos dos fármacos , Espectrina/análise
9.
Oncol Rep ; 11(1): 161-5, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14654920

RESUMO

In this work we studied the effect of v-raf and different form RAF on up regulation of VEGF gene expression and angiogenesis. In the experiments the truncated forms of RAF1 gene were transfected into the cells of the established human urothelial line HCV-29, otherwise non-angiogenic. The plasmids coding for mitochondrial targeted dominant active or dominant negative RAF1 (DAM, DNM respectively) were deleted with RAS binding domain. DAM has the ability to phosphorylate BAD whereas DNM cannot phosphorylate BAD. DAM RAF1, similarly to v-raf transfected cells, induced up-regulation of VEGF and caused angiogenic phenotype as studied in vivo. DNM RAF1 transfectants induced VEGF independent angiogenesis.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas c-raf/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/administração & dosagem , Plasmídeos/genética , Proteínas Proto-Oncogênicas c-raf/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Transcrição Gênica/genética , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Cell Mol Biol Lett ; 7(4): 1047-57, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12511971

RESUMO

We studied the effect of the modification of focal adhesion kinase (FAK) on the growth, migration and adhesion of C3H 10T1/2 cells. Cells transfected with plasmid coding for antisense FAK displayed a low level of FAK protein. Interestingly, the transfected cells achieved a higher saturation density at confluence, and displayed reduced adhesion and enhanced emigration from a confluent layer of cells when stimulated with fibronectin. In conclusion, it can be postulated that FAK plays an important role in the mechanism of contact inhibition.


Assuntos
Movimento Celular/fisiologia , DNA Antissenso , DNA Complementar/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Bioensaio , Linhagem Celular , Regulação para Baixo , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...