Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Mol Med ; 27(17): 2572-2582, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37537749

RESUMO

Anti-cancer properties of (-)-epigallocatechin-3-gallate (EGCG) are mediated via apoptosis induction, as well as inhibition of cell proliferation and histone deacetylase. Accumulation of stabilized cellular FLICE-inhibitory protein (c-FLIP)/Ku70 complex in the cytoplasm inhibits apoptosis through interruption of extrinsic apoptosis pathway. In this study, we evaluated the anti-cancer role of EGCG in gastric cancer (GC) cells through dissociation of c-FLIP/Ku70 complex. MKN-45 cells were treated with EGCG or its antagonist MG149 for 24 h. Apoptosis was evaluated by flow cytometry and quantitative RT-PCR. Protein expression of c-FLIP and Ku70 was analysed using western blot and immunofluorescence. Dissociation of c-FLIP/Ku70 complex as well as Ku70 translocation were studied by sub-cellular fractionation and co-immunoprecipitation. EGCG induced apoptosis in MKN-45 cells with substantial up-regulation of P53 and P21, down-regulation of c-Myc and Cyclin D1 as well as cell cycle arrest in S and G2/M check points. Moreover, EGCG treatment suppressed the expression of c-FLIP and Ku70, decreased their interaction while increasing the Ku70 nuclear content. By dissociating the c-FLIP/Ku70 complex, EGCG could be an alternative component to the conventional HDAC inhibitors in order to induce apoptosis in GC cells. Thus, its combination with other cancer therapy protocols could result in a better therapeutic outcome.


Assuntos
Catequina , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamento farmacológico , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Apoptose , Catequina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células
2.
Cell Mol Life Sci ; 80(2): 44, 2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36652019

RESUMO

Preeclampsia is a pregnancy-specific cardiovascular disorder, involving significant maternal endothelial dysfunction. Although inappropriate placentation due to aberrant angiogenesis, inflammation and shallow trophoblast invasion are the root causes of preeclampsia, pathogenic mechanisms are poorly understood, particularly in early pregnancy. Here, we first confirm the abnormal expression of important vascular and inflammatory proteins, FK506-binding protein-like (FKBPL) and galectin-3 (Gal-3), in human plasma and placental tissues from women with preeclampsia and normotensive controls. We then employ a three-dimensional microfluidic placental model incorporating human umbilical vein endothelial cells (HUVECs) and a first trimester trophoblast cell line (ACH-3P) to investigate FKBPL and Gal-3 signaling in inflammatory conditions. In human samples, both circulating (n = 17 controls; n = 30 preeclampsia) and placental (n ≥ 6) FKBPL and Gal-3 levels were increased in preeclampsia compared to controls (plasma: FKBPL, p < 0.0001; Gal-3, p < 0.01; placenta: FKBPL, p < 0.05; Gal-3, p < 0.01), indicative of vascular dysfunction in preeclampsia. In our placenta-on-a-chip model, we show that endothelial cells are critical for trophoblast-mediated migration and that trophoblasts effectively remodel endothelial vascular networks. Inflammatory cytokine tumour necrosis factor-α (10 ng/mL) modulates both FKBPL and Gal-3 signaling in conjunction with trophoblast migration and impairs vascular network formation (p < 0.005). Our placenta-on-a-chip recapitulates aspects of inappropriate placental development and vascular dysfunction in preeclampsia.


Assuntos
Placenta , Pré-Eclâmpsia , Gravidez , Feminino , Humanos , Placenta/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Trofoblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteínas de Ciclo Celular/metabolismo , Dispositivos Lab-On-A-Chip , Proteínas de Ligação a Tacrolimo/metabolismo
3.
Cancer Sci ; 114(4): 1337-1352, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36479791

RESUMO

Hepatocellular carcinoma (HCC) is a heterogeneous, late-diagnosed, and highly recurrent malignancy that often affects the whole body's metabolism. Finding certain theranostic molecules that can address current concerns simultaneously is one of the priorities in HCC management. In this study, performing protein-protein interaction network analysis proposed hepatocyte nuclear factor 4 alpha (HNF4α) as a hub protein, associating epithelial-mesenchymal transition (EMT) to reprogrammed cancer metabolism, formerly known as the Warburg effect. Both phenomena improved the compensation of cancerous cells in competitive conditions. Mounting evidence has demonstrated that HNF4α is commonly downregulated and serves as a tumor suppressor in the HCC. Enhancing the HNF4α mRNA translation through a specific synthetic antisense long non-coding RNA, profoundly affects both EMT and onco-metabolic modules in HCC cells. HNF4α overexpression decreased featured mesenchymal transcription factors and improved hepatocytic function, decelerated glycolysis, accelerated gluconeogenesis, and improved dysregulated cholesterol metabolism. Moreover, HNF4α overexpression inhibited the migration, invasion, and proliferation of HCC cells and decreased metastasis rate and tumor growth in xenografted nude mice. Our findings suggest a central regulatory role for HNF4α through its broad access to a wide variety of gene promoters involved in EMT and the Warburg effect in human hepatocytes. This essential impact indicates that HNF4α may be a potential target for HCC treatment.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Camundongos , Animais , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Transição Epitelial-Mesenquimal/genética , Camundongos Nus , Linhagem Celular Tumoral , Recidiva Local de Neoplasia/genética , Fator 4 Nuclear de Hepatócito/genética , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética
4.
Front Genet ; 13: 938985, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35938032

RESUMO

Analysis of circulating tumor DNA (ctDNA) can be used to characterize and monitor cancers. Recently, non-invasive prenatal testing (NIPT) as a new next-generation sequencing (NGS)-based approach has been applied for detecting ctDNA. This study aimed to investigate the copy number variations (CNVs) utilizing the non-invasive prenatal testing in plasma ctDNA from ovarian cancer (OC) patients who were treated with neoadjuvant chemotherapy (NAC). The plasma samples of six patients, including stages II-IV, were collected during the pre- and post-NAC treatment that were divided into NAC-sensitive and NAC-resistant groups during the follow-up time. CNV analysis was performed using the NIPT via two methods "an open-source algorithm WISECONDORX and NextGENe software." Results of these methods were compared in pre- and post-NAC of OC patients. Finally, bioinformatics tools were used for data mining from The Cancer Genome Atlas (TCGA) to investigate CNVs in OC patients. WISECONDORX analysis indicated fewer CNV changes on chromosomes before treatment in the NAC-sensitive rather than NAC-resistant patients. NextGENe data indicated that CNVs are not only observed in the coding genes but also in non-coding genes. CNVs in six genes were identified, including HSF1, TMEM249, MROH1, GSTT2B, ABR, and NOMO2, only in NAC-resistant patients. The comparison of these six genes in NAC-resistant patients with The Cancer Genome Atlas data illustrated that the total alteration frequency is amplification, and the highest incidence of the CNVs (≥35% based on TCGA data) is found in MROH1, TMEM249, and HSF1 genes on the chromosome (Chr) 8. Based on TCGA data, survival analysis showed a significant reduction in the overall survival among chemotherapy-resistant patients as well as a high expression level of these three genes compared to that of sensitive samples (all, p < 0.0001). The continued Chr8 study using WISECONDORX revealed CNV modifications in NAC-resistant patients prior to NAC therapy, but no CNV changes were observed in NAC-sensitive individuals. Our findings showed that low coverage whole-genome sequencing analysis used for NIPT could identify CNVs in ctDNA of OC patients before and after chemotherapy. These CNVs are different in NAC-sensitive and -resistant patients highlighting the potential application of this approach in cancer patient management.

5.
J Cell Physiol ; 237(11): 3984-4000, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36037302

RESUMO

Development is a symphony of cells differentiation in which different signaling pathways are orchestrated at specific times and periods to form mature and functional cells from undifferentiated cells. The similarity of the gene expression profile in malignant and undifferentiated cells is an interesting topic that has been proposed for many years and gave rise to the differentiation-therapy concept, which appears a rational insight and should be reconsidered. Hepatocellular carcinoma (HCC), as the sixth common cancer and the third leading cause of cancer death worldwide, is one of the health-threatening complications in communities where hepatotropic viruses are endemic. Sedentary lifestyle and high intake of calories are other risk factors. HCC is a complex condition in which various dimensions must be addressed, including heterogeneity of cells in the tumor mass, high invasiveness, and underlying diseases that limit the treatment options. Under these restrictions, recognizing, and targeting common signaling pathways during liver development and HCC could expedite to a rational therapeutic approach, reprograming malignant cells to well-differentiated ones in a functional state. Accordingly, in this review, we highlighted the commonalities of signaling pathways in hepatogenesis and hepatocarcinogenesis, and comprised an update on the current status of targeting these pathways in laboratory studies and clinical trials.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Carcinogênese/genética , Carcinogênese/patologia , Transdução de Sinais
6.
Methods Mol Biol ; 2535: 49-57, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35867221

RESUMO

Breast cancer (BC) is a leading cause of cancer death among women worldwide. To better understand and predict therapeutic response in BC patient developing a fast, low-cost, and reliable preclinical tumor from patient's tumor specimen is needed. Here, we describe the development of a preclinical model of BC through the generation and ex vivo culture of patient-derived organotypic tumor spheroids (PDOTS) in a 3D microfluidic device. Moreover, the real-time screening of conventional chemotherapy agents on cultured PDOTS is also described.


Assuntos
Neoplasias da Mama , Técnicas Analíticas Microfluídicas , Neoplasias da Mama/tratamento farmacológico , Técnicas de Cultura de Células , Feminino , Humanos , Dispositivos Lab-On-A-Chip , Esferoides Celulares
7.
PLoS One ; 17(4): e0267291, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35476804

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are frequently deregulated in various types of cancer. While antisense oligonucleotides are used to block oncomiRs, delivery of tumour-suppressive miRNAs holds great potential as a potent anti-cancer strategy. Here, we aim to determine, and functionally analyse, miRNAs that are lowly expressed in various types of tumour but abundantly expressed in multiple normal tissues. METHODS: The miRNA sequencing data of 14 cancer types were downloaded from the TCGA dataset. Significant differences in miRNA expression between tumor and normal samples were calculated using limma package (R programming). An adjusted p value < 0.05 was used to compare normal versus tumor miRNA expression profiles. The predicted gene targets were obtained using TargetScan, miRanda, and miRDB and then subjected to gene ontology analysis using Enrichr. Only GO terms with an adjusted p < 0.05 were considered statistically significant. All data from wet-lab experiments (cell viability assays and flow cytometry) were expressed as means ± SEM, and their differences were analyzed using GraphPad Prism software (Student's t test, p < 0.05). RESULTS: By compiling all publicly available miRNA profiling data from The Cancer Genome Atlas (TCGA) Pan-Cancer Project, we reveal a small set of tumour-suppressing miRNAs (which we designate as 'normomiRs') that are highly expressed in 14 types of normal tissues but poorly expressed in corresponding tumour tissues. Interestingly, muscle-enriched miRNAs (e.g. miR-133a/b and miR-206) and miRNAs from DLK1-DIO3 locus (e.g. miR-381 and miR-411) constitute a large fraction of the normomiRs. Moreover, we define that the CCCGU motif is absent in the oncomiRs' seed sequences but present in a fraction of tumour-suppressive miRNAs. Finally, the gain of function of candidate normomiRs across several cancer cell types indicates that miR-206 and miR-381 exert the most potent inhibition on multiple cancer types in vitro. CONCLUSION: Our results reveal a pan-cancer set of tumour-suppressing miRNAs and highlight the potential of miRNA-replacement therapies for targeting multiple types of tumour.


Assuntos
MicroRNAs , Neoplasias , Bases de Dados Factuais , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias/genética
8.
Trends Biotechnol ; 40(9): 1041-1060, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35311650

RESUMO

Multimodal analysis of circulating tumour cells (CTCs) has the potential to provide remarkable insight for cancer development and metastasis. CTCs and CTC clusters investigation using single-cell analysis, enables researchers to gain crucial information on metastatic mechanisms and the genomic alterations responsible for drug resistance, empowering treatment, and management of cancer. Despite a plethora of CTC isolation technologies, careful attention to the strengths and weaknesses of each method should be considered in order to isolate these rare cells. Here, we provide an overview of cutting-edge technologies used for single-cell isolation and analysis of CTCs. Additionally, we highlight the biological features, clinical application, and the therapeutic potential of CTCs and CTC clusters using single-cell analysis platforms for cancer management.


Assuntos
Células Neoplásicas Circulantes , Separação Celular/métodos , Humanos , Análise de Célula Única
9.
Theranostics ; 11(16): 8057-8075, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335980

RESUMO

Over the last few years, immunotherapy, in particular, immune checkpoint inhibitor therapy, has revolutionized the treatment of several types of cancer. At the same time, the uptake in clinical oncology has been slow owing to the high cost of treatment, associated toxicity profiles and variability of the response to treatment between patients. In response, personalized approaches based on predictive biomarkers have emerged as new tools for patient stratification to achieve effective immunotherapy. Recently, the enumeration and molecular analysis of circulating tumor cells (CTCs) have been highlighted as prognostic biomarkers for the management of cancer patients during chemotherapy and for targeted therapy in a personalized manner. The expression of immune checkpoints on CTCs has been reported in a number of solid tumor types and has provided new insight into cancer immunotherapy management. In this review, we discuss recent advances in the identification of immune checkpoints using CTCs and shed light on the potential applications of CTCs towards the identification of predictive biomarkers for immunotherapy.


Assuntos
Imunoterapia/métodos , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/metabolismo , Humanos , Fatores Imunológicos , Neoplasias/imunologia , Neoplasias/terapia , Medicina de Precisão/métodos , Prognóstico
10.
J Cell Mol Med ; 25(18): 8602-8614, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34423899

RESUMO

Hepatocellular carcinoma (HCC), the most common type of liver cancer, is usually a latent and asymptomatic malignancy caused by different aetiologies, which is a result of various aberrant molecular heterogeneity and often diagnosed at advanced stages. The incidence and prevalence have significantly increased because of sedentary lifestyle, diabetes, chronic infection with hepatotropic viruses and exposure to aflatoxins. Due to advanced intra- or extrahepatic metastasis, recurrence is very common even after radical resection. In this paper, we highlighted novel therapeutic modalities, such as molecular-targeted therapies, targeted radionuclide therapies and epigenetic modification-based therapies. These topics are trending headlines and their combination with cell-based immunotherapies, and gene therapy has provided promising prospects for the future of HCC treatment. Moreover, a comprehensive overview of current and advanced therapeutic approaches is discussed and the advantages and limitations of each strategy are described. Finally, very recent and approved novel combined therapies and their promising results in HCC treatment have been introduced.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Combinada/métodos , Imunoterapia/métodos , Neoplasias Hepáticas/terapia , Terapia de Alvo Molecular/métodos , Animais , Humanos
11.
Biosens Bioelectron ; 175: 112833, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33288428

RESUMO

Cell co-culture serves as a standard method to study intercellular communication. However, random diffusion of signal molecules during co-culture may arouse crosstalk among different types of cells and hide directive signal-target responses. Here, a microfluidic chip is proposed to study unidirectional intercellular communication by spatially controlling the flow of the signal molecules. The chip contains two separated chambers connected by two channels where the culture media flows oppositely. A zigzag signal-blocking channel is designed to study the function of a specific signal. The chip is applied to study the unidirectional communication between tumor cells and stromal cells. It shows that the expression of α-smooth muscle actin (a marker of cancer-associated fibroblast (CAF)) of both MRC-5 fibroblasts and mesenchymal stem cells can be up-regulated only by the secreta from invasive MDA-MB-231 cells, but not from non-invasive MCF-7 cells. The proliferation of the tumor cells can be improved by the stromal cells. Moreover, transforming growth factor beta 1 is found as one of the main factors for CAF transformation via the signal-blocking function. The chip achieves unidirectional cell communication along X-axis, signal concentration gradient along Y-axis and 3D cell culture along Z-axis, which provides a useful tool for cell communication studies.


Assuntos
Técnicas Biossensoriais , Microfluídica , Comunicação Celular , Técnicas de Cocultura , Fibroblastos , Humanos
12.
J Cell Physiol ; 236(5): 3918-3928, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33145762

RESUMO

Various factors in the tumor microenvironment (TME) regulate the expression of PD-L1 in cancer cells. In TME, mesenchymal stem cells (MSCs) play a crucial role in tumor progression, metastasis, and drug resistance. Emerging evidence suggests that MSCs can modulate the immune-suppression capacity of TME through the stimulation of PD-L1 expression in various cancers; nonetheless, their role in the induction of PD-L1 in breast cancer remained elusive. Here, we assessed the potential of MSCs in the stimulation of PD-L1 expression in a low PD-L1 breast cancer cell line and explored its associated cytokine. We assessed the expression of MSCs-related genes and their correlation with PD-L1 across 1826 breast cancer patients from the METABRIC cohort. After culturing an ER+/differentiated/low PD-L1 breast cancer cells with MSCs conditioned-medium (MSC-CM) in a microfluidic device, a variety of in-vitro assays was carried out to determine the role of MSC-CM in breast cancer cells' phenotype plasticity, invasion, and its effects on induction of PD-L1 expression. In-silico analysis showed a positive association between MSCs-related genes and PD-L1 expression in various types of breast cancer. Through functional assays, we revealed that MSC-CM not only prompts a phenotype switch but also stimulates PD-L1 expression at the protein level through secretion of various cytokines, especially CCL5. Treatment of MSCs with cytokine inhibitor pirfenidone showed a significant reduction in the secretion of CCL5 and consequently, expression of PD-L1 in breast cancer cells. We concluded that MSCs-derived CCL5 may act as a PD-L1 stimulator in breast cancer.


Assuntos
Antígeno B7-H1/metabolismo , Quimiocina CCL5/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Terapia de Imunossupressão , Células MCF-7 , Invasividade Neoplásica , Estadiamento de Neoplasias
13.
J Ovarian Res ; 13(1): 124, 2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-33076944

RESUMO

Ovarian cancer is the eighth most commonly occurring cancer in women. Clinically, the limitation of conventional screening and monitoring approaches inhibits high throughput analysis of the tumor molecular markers toward prediction of treatment response. Recently, analysis of liquid biopsies including circulating tumor DNA (ctDNA) open new way toward cancer diagnosis and treatment in a personalized manner in various types of solid tumors. In the case of ovarian carcinoma, growing pre-clinical and clinical studies underscored promising application of ctDNA in diagnosis, prognosis, and prediction of treatment response. In this review, we accumulate and highlight recent molecular findings of ctDNA analysis and its associations with treatment response and patient outcome. Additionally, we discussed the potential application of ctDNA in the personalized treatment of ovarian carcinoma. ctDNA-monitoring usage during the ovarian cancer treatments procedures.


Assuntos
DNA Tumoral Circulante/genética , Neoplasias Ovarianas/genética , Feminino , Humanos , Neoplasias Ovarianas/patologia , Prognóstico
14.
Adv Clin Chem ; 99: 193-235, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32951637

RESUMO

Cancer is a heterogeneous disease that requires a multimodal approach to diagnose, manage and treat. A better understanding of the disease biology can lead to identification of novel diagnostic/prognostic biomarkers and the discovery of the novel therapeutics with the goal of improving patient outcomes. Employing advanced technologies can facilitate this, enabling better diagnostic and treatment for cancer patients. In this regard, microfluidic technology has emerged as a promising tool in the studies of cancer, including single cancer cell analysis, modeling angiogenesis and metastasis, drug screening and liquid biopsy. Microfluidic technologies have opened new ways to study tumors in the preclinical and clinical settings. In this chapter, we highlight novel application of this technology in area of fundamental, translational and clinical cancer research.


Assuntos
Técnicas Analíticas Microfluídicas/métodos , Neoplasias/patologia , Neoplasias/terapia , Animais , Movimento Celular , Ensaios de Seleção de Medicamentos Antitumorais/instrumentação , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Desenho de Equipamento , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Invasividade Neoplásica/diagnóstico , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Neoplasias/diagnóstico , Neovascularização Patológica/diagnóstico , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Medicina de Precisão/instrumentação , Medicina de Precisão/métodos , Análise de Célula Única/instrumentação , Análise de Célula Única/métodos
15.
Integr Biol (Camb) ; 12(7): 188-197, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32638026

RESUMO

Various factors in the tumor microenvironment (TME) regulate the expression of PD-L1 in carcinoma cells. The cancer-associated fibroblasts (CAFs) play a crucial role in regulating and rewiring TME to enhance their immune suppressive function and to favor the invasion of the malignant cells. Tumor progression may be retarded by targeting CAFs in the TME. Various studies highlighted the ability of targeting CAF with pirfenidone (PFD), leading to increased efficacy of chemotherapy. However, its potential for the reduction of immune-suppression capacity of CAFs remains to be elusive. Here, we assessed the effect of PFD on the expression of PD-L1 on CAF cells. Besides migration inhibitory effects of PFD on CAFs, the expression level of PD-L1 reduced in CAFs after treatment with PFD. The downstream analysis of released cytokines from CAFs showed that PFD significantly dropped the secretion of CCL17 and TNF-ß, where a positive association between PFD-targeted proteins and PD-L1 was observed. These data suggest that the treatment of CAF within TME through the PFD may reduce the acquisition of CAF-mediated invasive and immune-suppressive capacity of breast carcinoma cells.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Neoplasias da Mama/imunologia , Fibroblastos Associados a Câncer/efeitos dos fármacos , Quimiocina CCL17/metabolismo , Terapia de Imunossupressão , Linfotoxina-alfa/metabolismo , Piridonas/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Piridonas/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos
16.
Crit Rev Biotechnol ; 40(2): 213-230, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31906727

RESUMO

Recently, organ-on-a-chip models, which are microfluidic devices that mimic the cellular architecture and physiological environment of an organ, have been developed and extensively investigated. The chips can be tailored to accommodate the disease conditions pertaining to many organs; and in the case of this review, the lung. Lung-on-a-chip models result in a more accurate reflection compared to conventional in vitro models. Pharmaceutical drug testing methods traditionally use animal models in order to evaluate pharmacological and toxicological responses to a new agent. However, these responses do not directly reflect human physiological responses. In this review, current and future applications of the lung-on-a-chip in the respiratory system will be discussed. Furthermore, the limitations of current conventional in vitro models used for respiratory disease modeling and drug development will be addressed. Highlights of additional translational aspects of the lung-on-a-chip will be discussed in order to demonstrate the importance of this subject for medical research.


Assuntos
Dispositivos Lab-On-A-Chip , Doenças Respiratórias/fisiopatologia , Animais , Pesquisa Biomédica , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiologia , Modelos Biológicos , Fenômenos Farmacológicos e Toxicológicos , Impressão Tridimensional , Doenças Respiratórias/tratamento farmacológico , Engenharia Tecidual
17.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118526, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31398408

RESUMO

Expression of programmed death-ligand 1 (PD-L1) in cancer cells plays an important role in cancer-immune cell interaction. The emerging evidence suggests regulation of PD-L1 expression by several tumor microenvironmental cues. However, the association of PD-L1 expression with chemical and mechanical features of the tumor microenvironment, specifically epidermal growth factor receptor (EGFR) signaling and matrix stiffness, remains elusive. Herein, we determine whether EGFR targeting and substrate stiffness affect the regulation of PD-L1 expression. Breast carcinoma cell lines, MCF7 and MDA-MB-231, were cultured under different conditions targeting EGFR and exposing cells to distinct substrate stiffness to evaluate PD-L1 expression. Furthermore, the ability to form aggregates in short-term culture of breast carcinoma cells and its effect on expression level of PD-L1 was probed. Our results indicated that PD-L1 expression was altered in response to both EGFR inhibition and substrate stiffness. Additionally, a positive association between the formation of multicellular aggregates and PD-L1 expression was observed. MDA-MB-231 cells expressed the highest PD-L1 level on a stiff substrate, while inhibition of EGFR reduced expression of PD-L1. The results suggested that both physical and chemical features of tumor microenvironment regulate PD-L1 expression through alteration of tumor aggregate formation potential. In line with these results, the in-silico study highlighted a positive correlation between PD-L1 expression, EGFR signaling, epithelial to mesenchymal transition related transcription factors (EMT-TFs) and stemness markers in metastatic breast cancer. These findings improve our understanding of regulation of PD-L1 expression by tumor microenvironment leading to evasion of tumor cells from the immune system.


Assuntos
Antígeno B7-H1/biossíntese , Neoplasias da Mama/metabolismo , Regulação para Cima , Neoplasias da Mama/patologia , Receptores ErbB/metabolismo , Feminino , Humanos , Células MCF-7 , Transdução de Sinais , Células Tumorais Cultivadas , Microambiente Tumoral
18.
Trends Biotechnol ; 36(4): 358-371, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29366522

RESUMO

Personalized cancer therapy applies specific treatments to each patient. Using personalized tumor models with similar characteristics to the original tumors may result in more accurate predictions of drug responses in patients. Tumor organoid models have several advantages over pre-existing models, including conserving the molecular and cellular composition of the original tumor. These advantages highlight the tremendous potential of tumor organoids in personalized cancer therapy, particularly preclinical drug screening and predicting patient responses to selected treatment regimens. Here, we highlight the advantages, challenges, and translational potential of tumor organoids in personalized cancer therapy and focus on gene-drug associations, drug response prediction, and treatment selection. Finally, we discuss how microfluidic technology can contribute to immunotherapy drug screening in tumor organoids.


Assuntos
Neoplasias/terapia , Organoides/transplante , Medicina de Precisão , Animais , Técnicas de Cultura de Células , Heterogeneidade Genética , Xenoenxertos/imunologia , Xenoenxertos/transplante , Humanos , Sistema Imunitário/imunologia , Imunoterapia , Camundongos , Microfluídica , Modelos Animais , Neoplasias/genética , Neoplasias/imunologia , Organoides/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...