Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.573
Filtrar
1.
Bull Cancer ; 109(1): 58-64, 2022 Jan.
Artigo em Francês | MEDLINE | ID: mdl-34903368

RESUMO

Three-dimensional (3D) culture of organoids from primary cells (wild type) or tumoroids from tumor cells, is used to study the physiological mechanisms in vivo, in order to model normal or tumor tissues more accurately than conventional two-dimensional (2D) culture. The features of this 3D culture, such as the three-dimensional structure, the self-renewal capacity and differentiation are preserved and appropriate to cancer study since their cellular characteristics are very similar to in vivo models. Here, we summarize the recent advances in the rapidly evolving field of organoids and their applications to cancer biology, clinical research and personalized medicine.


Assuntos
Pesquisa Biomédica , Técnicas de Cultura de Células em Três Dimensões/métodos , Neoplasias/patologia , Organoides/patologia , Brônquios/anatomia & histologia , Carcinogênese/genética , Diferenciação Celular , Autorrenovação Celular , Humanos , Neoplasias Pulmonares/patologia , Neoplasias/genética , Medicina de Precisão , Células Tumorais Cultivadas/patologia
2.
FEBS J ; 289(5): 1302-1314, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34036737

RESUMO

Adaptation of cellular function with the nutrient environment is essential for survival. Failure to adapt can lead to cell death and/or disease. Indeed, energy metabolism alterations are a major contributing factor for many pathologies, including cancer, cardiovascular disease, and diabetes. In particular, a primary characteristic of cancer cells is altered metabolism that promotes survival and proliferation even in the presence of limited nutrients. Interestingly, recent studies demonstrate that metabolic pathways produce intermediary metabolites that directly influence epigenetic modifications in the genome. Emerging evidence demonstrates that metabolic processes in cancer cells fuel malignant growth, in part, through epigenetic regulation of gene expression programs important for proliferation and adaptive survival. In this review, recent progress toward understanding the relationship of cancer cell metabolism, epigenetic modification, and transcriptional regulation will be discussed. Specifically, the need for adaptive cell metabolism and its modulation in cancer cells will be introduced. Current knowledge on the emerging field of metabolite production and epigenetic modification will also be reviewed. Alterations of DNA (de)methylation, histone modifications, such as (de)methylation and (de)acylation, as well as chromatin remodeling, will be discussed in the context of cancer cell metabolism. Finally, how these epigenetic alterations contribute to cancer cell phenotypes will be summarized. Collectively, these studies reveal that both metabolic and epigenetic pathways in cancer cells are closely linked, representing multiple opportunities to therapeutically target the unique features of malignant growth.


Assuntos
Carcinogênese/genética , Epigênese Genética , Histonas/genética , Neoplasias/genética , Transcrição Gênica , Células Tumorais Cultivadas/metabolismo , Antineoplásicos/uso terapêutico , Carcinogênese/metabolismo , Carcinogênese/patologia , Proliferação de Células/efeitos dos fármacos , Cromatina/química , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Histonas/antagonistas & inibidores , Histonas/metabolismo , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
3.
Oncol Rep ; 46(6)2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34651665

RESUMO

Breast cancer cell lines are widely used as an in vitro system with which to study the mechanisms underlying biological and chemotherapeutic resistance. In the present study, two novel breast cancer cell lines designated as PC­B­142CA and PC­B­148CA were successfully established from HER2­positive and triple­negative (TN) breast cancer tissues. The cell lines were characterized by cytokeratin (CK), α­smooth muscle actin (α­SMA), fibroblast­activation protein (FAP) and programmed death­ligand 1 (PD­L1). Cell proliferation was assessed using a colony formation assay, an MTS assay, 3­dimensional (3­D) spheroid and 3­D organoid models. Wound healing and Transwell migration assays were used to explore the cell migration capability. The responses to doxorubicin (DOX) and paclitaxel (PTX) were evaluated by 3­D spheroids. The results showed that the PC­B­142CA and PC­B­148CA cell lines were α­SMA­negative, FAP­negative, CK­positive and PD­L1­positive. Both cell lines were adherent with the ability of 3­D­multicellular spheroid and organoid formations; invadopodia were found in the spheroids/organoids of only PC­B­148CA. PC­B­142CA had a faster proliferative but lower metastatic rate compared to PC­B­148CA. Compared to MDA­MB­231, a commercial TN breast cancer cell line, PC­B­148CA had a similar CD44+/CD24­ stemness property (96.90%), whereas only 8.75% were found in PC­B­142CA. The mutations of BRCA1/2, KIT, PIK3CA, SMAD4, and TP53 were found in PC­B­142CA cells related to the resistance of several drugs, whereas PC­B­148CA had mutated BRCA2, NRAS and TP53. In conclusion, PC­B­142CA can serve as a novel HER2­positive breast cancer cell line for drug resistance studies; while PC­B­148CA is a novel TN breast cancer cell line suitable for metastatic and stemness­related properties.


Assuntos
Linhagem Celular Tumoral/patologia , Fragmentos de Peptídeos , Receptor ErbB-2 , Neoplasias de Mama Triplo Negativas/patologia , Movimento Celular , Proliferação de Células , Doxorrubicina/farmacologia , Feminino , Humanos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Organoides/patologia , Paclitaxel/farmacologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas/patologia
4.
Cancer Med ; 10(16): 5589-5598, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34240815

RESUMO

Patient-derived cancer models that reconstitute the characteristics of the tumor microenvironment may facilitate efforts in precision immune-oncology and the discovery of effective anticancer therapies. Organoids that have recently emerged as robust preclinical models typically contain tumor epithelial cells and lack the native tumor immune microenvironment. A patient-derived organotypic tumor spheroid (PDOTS) is a novel and innovative ex vivo system that retains key features of the native tumor immune microenvironment. Here, we established and characterized a series of colorectal cancer PDOTS models for use as a preclinical platform for testing effective immunotherapy and its combinations with other drugs. Partially dissociated (> 100 µm in diameter) tumor tissues were embedded in Matrigel-containing organoid media and subsequently formed into organoid structures within 3 to 7 days of culture. The success rate of growing PDOTS from fresh tissues was ~86%. Morphological analysis showed that the PDOTSs varied in size and structure. Immunofluorescence and flow cytometry analysis revealed that the PDOTSs retained autologous tumor-infiltrating lymphoid cells and tumor-infiltrating lymphoid cells were continually decreased through serial passages. Notably, PDOTSs from tumors from a high-level microsatellite instability-harboring patient were sensitive to anti-PD-1 or anti-PD-L1 antibodies. Our results demonstrate that the PDOTS model in which the tumor immune microenvironment is preserved may represent an advantageous ex vivo system to develop effective immune therapeutics.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Inibidores de Checkpoint Imunológico/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Instabilidade de Microssatélites , Cultura Primária de Células/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/imunologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/patologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
5.
STAR Protoc ; 2(2): 100409, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33870218

RESUMO

This protocol provides the procedures for isolating differentiated tumor cells from medulloblastoma (MB) in mice. Procedures for transplantation into cerebella are also included to examine the tumorigenesis of differentiated MB cells. This protocol outlines the detailed steps required for (1) isolation of tumor cells from mouse MB, (2) purification of differentiated tumor cells by fluorescence-activated cell sorting, and (3) transplantation of tumor cells into cerebella. This protocol is useful to purify differentiated tumor cells for investigating mechanisms underlying MB progression. For complete details on the use and execution of this protocol, please refer to Cheng et. al. (2020).


Assuntos
Separação Celular/métodos , Neoplasias Cerebelares/patologia , Cerebelo/cirurgia , Meduloblastoma/patologia , Células Tumorais Cultivadas , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Transplante de Células , Camundongos , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/transplante
6.
Adv Drug Deliv Rev ; 175: 113760, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33838208

RESUMO

The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.


Assuntos
Nanomedicina/métodos , Tecidos Suporte , Células Tumorais Cultivadas/patologia , Animais , Antineoplásicos/uso terapêutico , Humanos , Hidrogéis , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/terapia , Esferoides Celulares/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
7.
Pharmacol Ther ; 218: 107668, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32853629

RESUMO

Tumor development and progression require chemical and mechanical cues derived from cellular and non-cellular components in the tumor microenvironment, including the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), endothelial cells, and immune cells. Therefore, it is crucial to develop tissue culture models that can mimic in vivo cancer cell-ECM and cancer-stromal cell interactions. Three-dimensional (3D) tumor culture models have been widely utilized to study cancer development and progression. A recent advance in 3D culture is the development of patient-derived tumor organoid (PDO) models from primary human cancer tissue. PDOs maintain the heterogeneity of the primary tumor, which makes them more relevant for identifying therapeutic targets and verifying drug response. Other significant advances include development of 3D co-culture assays to investigate cell-cell interactions and tissue/organ morphogenesis, and microfluidic technology that can be integrated into 3D culture to mimic vasculature and blood flow. These advances offer spatial and temporal insights into cancer cell-stromal interactions and represent novel techniques to study tumor progression and drug response. Here, we summarize the recent progress in 3D culture and tumor organoid models, and discuss future directions and the potential of utilizing these models to study cancer-stromal interactions and direct personalized treatment.


Assuntos
Neoplasias , Medicina de Precisão , Comunicação Celular , Humanos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Organoides/patologia , Células Estromais/patologia , Células Tumorais Cultivadas/patologia , Microambiente Tumoral
8.
STAR Protoc ; 1(2)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-33043307

RESUMO

Tumor organoids are promising tools for cancer biology investigations and preclinical drug screenings because they are often representative of the histology and drug responses of patients. Here, we introduce a facile protocol to overcome technical limitations by generating patient-derived tumor organoids using a simplified ring-like geometry. This facilitates media exchange and drug treatment for histopathology characterization and automated high-throughput drug screenings. For complete details on the use and execution of this protocol, please refer to Phan et al. (2019).


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Histocitoquímica/métodos , Neoplasias , Organoides , Células Tumorais Cultivadas , Humanos , Neoplasias/química , Neoplasias/patologia , Organoides/citologia , Organoides/patologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/patologia
9.
Gynecol Oncol ; 159(2): 563-572, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958270

RESUMO

BACKGROUND: High fatality in ovarian cancer is attributed to metastasis, propagated by the release of multi-cellular aggregates/spheroids into the peritoneal cavity and their subsequent mesothelial invasion of peritoneal organs. Spheroids are therefore a common and clinically relevant in vitro model for ovarian cancer research. Spheroids in patients vary significantly in size and shape and display enhanced resistance to anti-cancer drugs compared to monolayers. However, there is no consensus on how spheroid size and shape affect drug resistance. Moreover, existing data regarding the influence of epithelial-to-mesenchymal transition (EMT) profile on spheroid shape and migration is inconclusive. METHODS: We formed spheroids with OVCAR-3 and OVCAR-8 cells, chosen for their established genetic similarity to the patient tumor samples. We monitored their morphology using confocal microscope with dipping objective and fluorescent microscope. We characterized important EMT biomarkers; E-cadherin, Vimentin and Slug through western blotting in monolayers and spheroids. We treated these spheroids with Taxol and Cisplatin and investigated their migratory profile based on their morphology. RESULTS: We report two distinct multicellular structures: loose aggregates (OVCAR-3) and compact spheroids (OVCAR-8). We attribute these different morphologies to the expression of the EMT biomarkers, and their changes upon spheroid formation. Importantly, we did not observe a difference in resistance to the anti-cancer drugs as a function of spheroid size and shape. However, migration capacity of compact spheroid (OVCAR-8) was 15-fold higher compared to that of loose aggregates (OVCAR-3). CONCLUSIONS: These results highlight the importance of spheroid size and shape on anti-cancer drug resistance and migration profiles. The results of this study can, therefore, help to elucidate general rules for ovarian cancer studies based on 3D samples.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Epitelial do Ovário/tratamento farmacológico , Cisplatino/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Carcinoma Epitelial do Ovário/genética , Movimento Celular , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Ovarianas/genética , Esferoides Celulares/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
10.
BMC Cancer ; 19(1): 628, 2019 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-31238897

RESUMO

BACKGROUND: A major barrier to effective treatment of glioblastoma (GBM) is the large intertumoral heterogeneity at the genetic and cellular level. In early phase clinical trials, patient heterogeneity in response to therapy is commonly observed; however, how tumor heterogeneity is reflected in individual drug sensitivities in the treatment-naïve glioblastoma stem cells (GSC) is unclear. METHODS: We cultured 12 patient-derived primary GBMs as tumorspheres and validated tumor stem cell properties by functional assays. Using automated high-throughput screening (HTS), we evaluated sensitivity to 461 anticancer drugs in a collection covering most FDA-approved anticancer drugs and investigational compounds with a broad range of molecular targets. Statistical analyses were performed using one-way ANOVA and Spearman correlation. RESULTS: Although tumor stem cell properties were confirmed in GSC cultures, their in vitro and in vivo morphology and behavior displayed considerable tumor-to-tumor variability. Drug screening revealed significant differences in the sensitivity to anticancer drugs (p < 0.0001). The patient-specific vulnerabilities to anticancer drugs displayed a heterogeneous pattern. They represented a variety of mechanistic drug classes, including apoptotic modulators, conventional chemotherapies, and inhibitors of histone deacetylases, heat shock proteins, proteasomes and different kinases. However, the individual GSC cultures displayed high biological consistency in drug sensitivity patterns within a class of drugs. An independent laboratory confirmed individual drug responses. CONCLUSIONS: This study demonstrates that patient-derived and treatment-naïve GSC cultures maintain patient-specific traits and display intertumoral heterogeneity in drug sensitivity to anticancer drugs. The heterogeneity in patient-specific drug responses highlights the difficulty in applying targeted treatment strategies at the population level to GBM patients. However, HTS can be applied to uncover patient-specific drug sensitivities for functional precision medicine.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Ensaios de Triagem em Larga Escala , Células-Tronco Neoplásicas/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos SCID , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Esferoides Celulares/patologia , Células Tumorais Cultivadas/patologia
11.
Biomaterials ; 194: 161-170, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30605824

RESUMO

Artificial stimuli-responsive hydrogels that can mimic natural extracellular matrix for growth and release of cancer spheroids (CSs) have attracted much attention. However, such hydrogels still face a challenge in regulating CSs growth and controlled release as well as keeping CSs integrity. Herein, a new class of ClO-/SCN- reversibly responsive nanocellulose hydrogel with fluorescence on-off reporter is developed. Upon addition of ClO-, the gel network of nanocellulose hydrogel was destructed, accompanying by the fluorescent quenching. Notably, when introducing of SCN-, a red fluorescence filamentous hydrogel was recovered by coordination cross-linking. The hydrogel reforms in a completely reversible process through the regulation of ClO-/SCN-. Benefit from the above response features of the hydrogel, the growth of cancer spheroids (CSs) in the hydrogel and on demand release of CSs from the hydrogel could be easily achieved through ClO-/SCN- regulation. Importantly, the growth and release of CSs can be monitored in real time by fluorescence imaging. Overall, such design strategy based on ClO-/SCN--responsive fluorescent hydrogels provided a new type of multi-responsive hydrogels as main scaffolds for cancer research and cancer drug screening.


Assuntos
Celulose/química , Hidrogéis/química , Substâncias Luminescentes/química , Esferoides Celulares/citologia , Ânions/química , Materiais Biocompatíveis/química , Técnicas de Cultura de Células , Humanos , Células MCF-7 , Neoplasias/patologia , Imagem Óptica , Esferoides Celulares/patologia , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/patologia
12.
Biomaterials ; 197: 296-304, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30682644

RESUMO

Mechanistic analysis of metastatic prostate cancer (PCa) biology and therapy response critically depends upon clinically relevant three-dimensional (3D) bone-like, organotypic culture. We here combine an engineered bone-mimetic environment (BME) with longitudinal microscopy to test the growth and therapy response of 3D PCa tumoroids. Besides promoting both tumor-cell autonomous and microenvironment-dependent growth in PCa cell lines and patient-derived xenograft cells, the BME enables in vivo-like tumor cell response to therapy, and reveals bone stroma dependent resistance to chemotherapy and BME-targeted localization and induction of cytoxicity by Radium-223. The BME platform will allow the propagation, compound screening and mechanistic dissection of patient-derived bone tumor isolates and applications toward personalized medicine.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Neoplasias da Próstata/tratamento farmacológico , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Técnicas de Cultura de Células/métodos , Proliferação de Células/efeitos dos fármacos , Docetaxel/farmacologia , Humanos , Masculino , Camundongos SCID , Células PC-3 , Neoplasias da Próstata/patologia , Rádio (Elemento)/farmacologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia , Microambiente Tumoral/efeitos dos fármacos
13.
Proc Natl Acad Sci U S A ; 115(50): E11671-E11680, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30487219

RESUMO

Tumorigenesis depends on intricate interactions between genetically altered tumor cells and their surrounding microenvironment. While oncogenic drivers in lung squamous carcinoma (LUSC) have been described, the role of stroma in modulating tissue architecture, particularly cell polarity, remains unclear. Here, we report the establishment of a 3D coculture system of LUSC epithelial cells with cancer-associated fibroblasts (CAFs) and extracellular matrix that together capture key components of the tumor microenvironment (TME). Single LUSC epithelial cells develop into acinar-like structures with 0.02% efficiency, and addition of CAFs provides proper tumor-stromal interactions within an appropriate 3D architectural context. Using this model, we recapitulate key pathological changes during tumorigenesis, from hyperplasia to dysplasia and eventually invasion, in malignant LUSC spheroids that undergo phenotypic switching in response to cell intrinsic and extrinsic changes. Overexpression of SOX2 is sufficient to mediate the transition from hyperplasia to dysplasia in LUSC spheroids, while the presence of CAFs makes them invasive. Unexpectedly, CAFs suppress the activity of high SOX2 levels, restore hyperplasia, and enhance the formation of acinar-like structures. Taken together, these observations suggest that stromal factors can override cell intrinsic oncogenic changes in determining the disease phenotype, thus providing fundamental evidence for the existence of dynamic reciprocity between the nucleus and the TME of LUSC.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fatores de Transcrição SOXB1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/genética , Polaridade Celular , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Humanos , Hiperplasia , Neoplasias Pulmonares/genética , Modelos Biológicos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB1/genética , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Microambiente Tumoral/genética , Regulação para Cima
14.
J Math Biol ; 77(4): 1073-1092, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29736873

RESUMO

Biological tissues accumulate mechanical stress during their growth. The mere measurement of the stored stress is not an easy task. We address here the spherical case and our experiments consist in performing an incision of a spherical microtissue (tumor spheroid) grown in vitro. On the theoretical part we derive a compatibility condition on the stored stress in spherical symmetry, which imposes a relation between the circumferential and radial stored stress. The numerical implementation uses the hyperelastic model of Ciarlet and Geymonat. A parametric study is performed to assess the influence of each parameter on the shape of the domain after the incision. As a conclusion, the total radial stored stress can be confidently estimated from the measurement of the opening after incision. We validate the approach with experimental data.


Assuntos
Modelos Biológicos , Neoplasias/patologia , Neoplasias/fisiopatologia , Fenômenos Biomecânicos , Simulação por Computador , Elasticidade , Células HCT116/patologia , Células HCT116/fisiologia , Humanos , Imageamento Tridimensional , Conceitos Matemáticos , Esferoides Celulares/patologia , Esferoides Celulares/fisiologia , Estresse Mecânico , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/fisiologia
15.
Stem Cell Res ; 27: 109-120, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29414601

RESUMO

BACKGROUND: Colorectal cancer (CRC) liver metastasis is highly unfavorable for patient outcome and is a leading cause of cancer-related death. Pre-clinical research of CRC liver metastasis predominately utilizes CRC cell lines grown in tissue culture. Here, we demonstrate that CRC liver metastases organoids derived from human specimens recapitulate some aspects of human disease. METHODS: Human CRC liver metastases pathological specimens were obtained following patient consent. Tumor disaggregates were plated and organoids were allowed to expand. CRC markers were identified by immunofluorescence. Stem cell genes were analysed by QPCR and flow cytometry. Response to drug therapy was quantified using time-lapse imaging and MATLAB analysis. RESULTS: Organoids showed global expression of the epithelial marker, EpCAM and the adenocarcinoma marker, CEA CAM1. Flow cytometry analysis demonstrated that organoids express the stem cell surface markers CD24 and CD44. Finally, we demonstrated that CRC liver metastases organoids acquire chemotherapy resistance and can be utilized as surrogates for drug testing. CONCLUSION: These data demonstrate that CRC liver metastases organoids recapitulate some aspects of human disease and may provide an invaluable resource for investigating novel drug therapies, chemotherapy resistance and mechanism of metastasis.


Assuntos
Neoplasias Colorretais/complicações , Neoplasias Hepáticas/secundário , Organoides/patologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Citometria de Fluxo , Humanos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia
16.
Sci Rep ; 7(1): 4575, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28676662

RESUMO

Human tumour progression is a dynamic process involving diverse biological and biochemical events such as genetic mutation and selection in addition to physical, chemical, and mechanical events occurring between cells and the tumour microenvironment. Using 3D bioprinting we have developed a method to embed MDA-MB-231 triple negative breast cancer cells, and IMR-90 fibroblast cells, within a cross-linked alginate/gelatin matrix at specific initial locations relative to each other. After 7 days of co-culture the MDA-MB-231 cells begin to form multicellular tumour spheroids (MCTS) that increase in size and frequency over time. After ~15 days the IMR-90 stromal fibroblast cells migrate through a non-cellularized region of the hydrogel matrix and infiltrate the MDA-MB-231 spheroids creating mixed MDA-MB-231/IMR-90 MCTS. This study provides a proof-of-concept that biomimetic in vitro tissue co-culture models bioprinted with both breast cancer cells and fibroblasts will result in MCTS that can be maintained for durations of several weeks.


Assuntos
Alginatos , Bioimpressão , Gelatina , Hidrogéis , Esferoides Celulares , Tecidos Suporte , Células Tumorais Cultivadas , Alginatos/química , Técnicas de Cocultura , Gelatina/química , Humanos , Hidrogéis/química , Neoplasias/patologia , Esferoides Celulares/patologia , Esferoides Celulares/ultraestrutura , Tecidos Suporte/química , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/ultraestrutura , Microambiente Tumoral
17.
Br J Haematol ; 178(5): 781-793, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28597546

RESUMO

New effective treatments are needed to improve outcomes for multiple myeloma (MM) patients. Receptors with restricted expression on plasma cells (PCs) represent attractive new therapeutic targets. The endothelin-1 (EDN1) axis, consisting of EDN1 acting through EDN-receptor A (EDNRA) and B (EDNRB), was previously shown to be overexpressed in several tumours, including MM. However, there is incomplete understanding of how EDN1 axis regulates MM growth and response to therapy. Besides EDNRA, the majority of MM cell lines and primary malignant PCs express high levels of EDNRB and release EDN1. Similarly, bone-marrow microenvironment cells also secrete EDN1. Investigating the extent of epigenetic dysregulation of EDNRB gene in MM, we found that hypermethylation of EDNRB promoter and subsequent down-regulation of EDNRB gene was observed in PCs or B lymphocytes from healthy donors compared to EDNRB-expressing malignant PCs. Pharmacological blockade with the dual EDN1 receptor antagonist bosentan decreased cell viability and MAPK activation of U266 and RPMI-8226 cells. Interestingly, the combination of bosentan and the proteasome inhibitor bortezomib, currently approved for MM treatment, resulted in synergistic cytotoxic effects. Overall, our data has uncovered EDN1-mediated autocrine and paracrine mechanisms that regulate malignant PCs growth and drug response, and support EDN1 receptors as new therapeutic targets in MM.


Assuntos
Antagonistas do Receptor de Endotelina A/farmacologia , Mieloma Múltiplo/sangue , Receptor de Endotelina A/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Comunicação Autócrina/fisiologia , Bortezomib/farmacologia , Bosentana , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Metilação de DNA , DNA de Neoplasias/genética , Sinergismo Farmacológico , Endotelina-1/sangue , Endotelina-1/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Plasmócitos/metabolismo , Regiões Promotoras Genéticas , Receptor de Endotelina A/genética , Sulfonamidas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
18.
Br J Haematol ; 178(6): 936-948, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28653353

RESUMO

The epigenome is often deregulated in cancer and treatment with inhibitors of bromodomain and extra-terminal proteins, the readers of epigenetic acetylation marks, represents a novel therapeutic approach. Here, we have characterized the anti-tumour activity of the novel bromodomain and extra-terminal (BET) inhibitor BAY 1238097 in preclinical lymphoma models. BAY 1238097 showed anti-proliferative activity in a large panel of lymphoma-derived cell lines, with a median 50% inhibitory concentration between 70 and 208 nmol/l. The compound showed strong anti-tumour efficacy in vivo as a single agent in two diffuse large B cell lymphoma models. Gene expression profiling showed BAY 1238097 targeted the NFKB/TLR/JAK/STAT signalling pathways, MYC and E2F1-regulated genes, cell cycle regulation and chromatin structure. The gene expression profiling signatures also highly overlapped with the signatures obtained with other BET Bromodomain inhibitors and partially overlapped with HDAC-inhibitors, mTOR inhibitors and demethylating agents. Notably, BAY 1238097 presented in vitro synergism with EZH2, mTOR and BTK inhibitors. In conclusion, the BET inhibitor BAY 1238097 presented promising anti-lymphoma preclinical activity in vitro and in vivo, mediated by the interference with biological processes driving the lymphoma cells. Our data also indicate the use of combination schemes targeting EZH2, mTOR and BTK alongside BET bromodomains.


Assuntos
Antineoplásicos/uso terapêutico , Benzodiazepinas/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Adenina/análogos & derivados , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Benzodiazepinas/administração & dosagem , Benzodiazepinas/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Everolimo/farmacologia , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/patologia , Camundongos SCID , Piperidinas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Pirimidinas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Handchir Mikrochir Plast Chir ; 49(2): 111-122, 2017 Apr.
Artigo em Alemão | MEDLINE | ID: mdl-28380641

RESUMO

Background Breast cancer is the world's most common cancer among women. Autologous lipotransfer is increasingly used for breast reconstruction following surgical removal of the tumour. In cell-assisted lipotransfer, the transplant is enriched with stem cells from adipose tissue (ADSC). Despite positive clinical results, there are some concerns regarding oncological safety due to transplanted stem cells. To date there are only a few breast cancer studies using primary cells from the same patient to enable further investigation into the complexity of cell-cell interactions in breast cancer in an experimental setting. Materials and methods We performed literature research on the topic of autologous lipotransfer. 5 different cell types (epithelial, mesenchymal cells, ADSC, endothelial cells, endothelial progenitor cells) were isolated from mammary (carcinoma) tissue or blood and were subsequently characterised for gene and protein expression as well as functional properties. The arteriovenous (AV) loop model in the rat was evaluated as a possible in vivo model for breast cancer pathogenesis and angiogenesis in this study. Results The literature provided evidence for an in-vitro interaction between ADSC and cells of the mammary (carcinoma) tissue. In some clinical studies, certain subgroups of patients appeared to be exposed to an increased risk of tumour recurrence after lipotransfer, but in most studies no correlation between lipotransfer and tumour recurrence was found. Different cell populations, which differed significantly in terms of surface markers, gene expression and functional properties, were isolated from tissue of the same patient. Axial vascularised tissue was successfully generated in the AV loop model. Conclusion In this study we were able to isolate different cell populations from the same patient, which reflect the heterogeneity of the tumour tissue. This enables a precise analysis of cell-cell interactions and their effects on tumour angiogenesis and pathogenesis in breast cancer. In combination with the AV loop model, this offers new possibilities to generate vascularised mammary carcinoma tissue as well as healthy mammary gland tissue in vivo as an optimal model for the clinical setting.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/irrigação sanguínea , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/irrigação sanguínea , Carcinoma Intraductal não Infiltrante/patologia , Carcinoma Lobular/irrigação sanguínea , Carcinoma Lobular/patologia , Separação Celular/métodos , Transformação Celular Neoplásica/patologia , Técnicas In Vitro , Transplante de Neoplasias/métodos , Transplante de Neoplasias/patologia , Neovascularização Patológica/patologia , Células Tumorais Cultivadas/patologia , Tecido Adiposo/patologia , Tecido Adiposo/transplante , Animais , Feminino , Humanos , Mamoplastia , Ratos , Retalhos Cirúrgicos/patologia , Transplante Autólogo
20.
Hepatology ; 65(2): 501-514, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27474881

RESUMO

The cancer microenvironment plays a central role in cancer development, growth, and homeostasis. This paradigm suggests that cancer fibroblasts support cancers, probably in response to stimuli received from the cancer cells. We aimed at investigating whether extracellular vesicles (EVs) can shuttle microRNA (miR) species between cancer-associated fibroblasts (CAFs) and cancer cells. To this end, we extracted EVs according to published protocols. EVs were studied for their miR content by quantitative reverse-transcription polymerase chain reaction. EVs were transfected with select miR species and utilized in vitro as well as in vivo in a rat model of cholangiocarcinoma (CCA). We found that miR-195 is down-regulated in CCA cells, as well as in adjoining fibroblasts. Furthermore, we report that EVs shuttle miR-195 from fibroblasts to cancer cells. Last, we show that fibroblast-derived EVs, loaded with miR-195, can be administered in a rat model of CCA, concentrate within the tumor, decrease the size of cancers, and improve survival of treated rats. CONCLUSION: EVs play a salient role in trafficking miR species between cancer cells and CAFs in human CCA. Understanding of these mechanisms may allow devising of novel therapeutics. (Hepatology 2017;65:501-514).


Assuntos
Neoplasias dos Ductos Biliares/mortalidade , Colangiocarcinoma/mortalidade , Vesículas Extracelulares/genética , MicroRNAs/farmacologia , Microambiente Tumoral/genética , Animais , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/patologia , Carcinogênese/genética , Movimento Celular/genética , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Modelos Animais de Doenças , Regulação para Baixo , Fibroblastos/patologia , Humanos , Imuno-Histoquímica , Masculino , MicroRNAs/genética , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sensibilidade e Especificidade , Taxa de Sobrevida , Transfecção , Células Tumorais Cultivadas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...