Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Lab Invest ; 102(2): 172-184, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34782726

RESUMO

The phenotype of glioma-initiating cells (GIC) is modulated by cell-intrinsic and cell-extrinsic factors. Phenotypic heterogeneity and plasticity of GIC is an important limitation to therapeutic approaches targeting cancer stem cells. Plasticity also presents a challenge to the identification, isolation, and propagation of purified cancer stem cells. Here we use a barcode labelling approach of GIC to generate clonal populations over a number of passages, in combination with phenotyping using the established stem cell markers CD133, CD15, CD44, and A2B5. Using two cell lines derived from isocitrate dehydrogenase (IDH)-wildtype glioblastoma, we identify a remarkable heterogeneity of the phenotypes between the cell lines. During passaging, clonal expansion manifests as the emergence of a limited number of barcoded clones and a decrease in the overall number of clones. Dual-labelled GIC are capable of forming traceable clonal populations which emerge after as few as two passages from mixed cultures and through analyses of similarity of relative proportions of 16 surface markers we were able to pinpoint the fate of such populations. By generating tumour organoids we observed a remarkable persistence of dominant clones but also a significant plasticity of stemness marker expression. Our study presents an experimental approach to simultaneously barcode and phenotype glioma-initiating cells to assess their functional properties, for example to screen newly established GIC for tumour-specific therapeutic vulnerabilities.


Assuntos
Antígenos CD/imunologia , Neoplasias Encefálicas/imunologia , Glioma/imunologia , Células-Tronco Neoplásicas/imunologia , Microambiente Tumoral/imunologia , Antígeno AC133/imunologia , Antígeno AC133/metabolismo , Antígenos CD/metabolismo , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Células Cultivadas , Células Clonais/imunologia , Células Clonais/metabolismo , Citometria de Fluxo , Glioma/metabolismo , Glioma/patologia , Humanos , Receptores de Hialuronatos/imunologia , Receptores de Hialuronatos/metabolismo , Imunofenotipagem , Antígenos CD15/imunologia , Antígenos CD15/metabolismo , Microscopia Confocal , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo
2.
Biochem Biophys Res Commun ; 558: 209-215, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32958251

RESUMO

Tumor heterogeneity is one of the ongoing huddles in the field of colon cancer therapy. It is evident that there are countless clones which exhibit different phenotypes and therefore, single cell analysis is inevitable. Cancer stem cells (CSCs) are rare cell population within tumor which is known to function in cancer metastasis and recurrence. Although there have been trials to prove intra-tumoral heterogeneity using single cell sequencing, that of CSCs has not been clearly elucidated. Here, we articulate the presence of heterogeneous subclones within CD133 positive cancer stem cells through single cell sequencing. As a proof of principle, we performed phenotype-based high-throughput laser isolation and single cell sequencing (PHLI-seq) of CD133 positive cells in a frozen tumor tissue obtained from a patient with colorectal cancer. The result proved that CD133 positive cells were shown to be heterogeneous both in copy number and mutational profiles. Single cancer stem cell specific mutations such as RNF144A, PAK2, PARP4, ADAM21, HYDIN, KRT38 and CELSR1 could be also detected in liver metastatic tumor of the same patient. Collectively, these data suggest that single cell analysis used to spot subclones with genetic variation within rare population, will lead to new strategies to tackle colon cancer metastasis.


Assuntos
Antígeno AC133/metabolismo , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo , Idoso , Biomarcadores Tumorais/metabolismo , Separação Celular/métodos , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Dosagem de Genes , Humanos , Lasers , Masculino , Mutação , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , Fenótipo , Análise de Célula Única , Sequenciamento do Exoma
3.
Hematol Oncol ; 37(4): 333-344, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30938848

RESUMO

Although the majority of patients with diffuse large B-cell lymphoma (DLBCL) can be cured with the standard immunochemotherapy R-CHOP, one-third of them relapses with a dismal outcome in most cases. In the recent years, remarkable advances have been achieved based on the discovery of molecular genetics in DLBCL. In addition to the major cell-of-origin designations of germinal center B-cell and activated B-cell subtypes, next-generation sequencing has unveiled the remarkable complexity of DLBCL and identified potential molecular targets for tailored therapies. Despite these findings, the current standard of care for DLBCL patients is still R-CHOP, and optimization of frontline therapy remains an important goal. In this review, we summarize recent updates on the evolution of frontline therapies for DLBCL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Terapia de Alvo Molecular , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Subpopulações de Linfócitos B/efeitos dos fármacos , Ensaios Clínicos como Assunto , Terapia Combinada , Quimioterapia de Consolidação , Ciclofosfamida/administração & dosagem , DNA de Neoplasias/genética , Doxorrubicina/administração & dosagem , Perfilação da Expressão Gênica , Genes Neoplásicos , Transplante de Células-Tronco Hematopoéticas , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/terapia , Quimioterapia de Manutenção , Estudos Multicêntricos como Assunto , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/efeitos dos fármacos , Prednisona/administração & dosagem , Intervalo Livre de Progressão , Ensaios Clínicos Controlados Aleatórios como Assunto , Indução de Remissão , Rituximab/administração & dosagem , Terapia de Salvação , Vincristina/administração & dosagem
4.
Biochem Biophys Res Commun ; 484(3): 631-635, 2017 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-28153736

RESUMO

To elucidate signaling pathways that regulate gastric cancer stem cell (CSC) phenotypes and immune checkpoint, we performed a proteogenomic analysis of NCC-S1M, which is a gastric cancer cell line with CSC-like characteristics and is the only syngeneic gastric tumor cell line transplant model created in the scientific community. We found that the NCC-S1M allograft was responsive to anti-PD-1 treatment, and overexpressed Cd274 encoding PD-L1. PD-L1 was transcriptionally activated by loss of the TGF-ß signaling. Il1rl1 protein was overexpressed in NCC-S1M cells compared with NCC-S1 cells that are less tumorigenic and less chemoresistant. Il1rl1 knockdown in NCC-S1M cells reduced tumorigenic potential and in vivo chemoresistance. Our proteogenomic analysis demonstrates a role of Smad4 loss in the PD-L1 immune evasion, as well as Il1rl1's role in CSC-like properties of NCC-S1M.


Assuntos
Proteínas de Neoplasias/imunologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Proteoma/imunologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/imunologia , Animais , Antineoplásicos , Carcinogênese/efeitos dos fármacos , Carcinogênese/imunologia , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/imunologia , Camundongos , Células-Tronco Neoplásicas/classificação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Neoplasias Gástricas/classificação
5.
J Calif Dent Assoc ; 44(2): 112-20, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26930754

RESUMO

The concept of tumor development driven by a unique subpopulation of cancer stem cells (CSCs), or the CSCs hypothesis, may help to explain the high mortality, low response to treatment and tendency of developing multiple tumors in oral cancer. We will review current knowledge of the CSCs hypothesis in oral cancer and the traits displayed by CSCs, focusing on the resistance to therapy and attempts being made to treat oral cancer by specifically targeting CSCs.


Assuntos
Neoplasias Bucais/patologia , Células-Tronco Neoplásicas/fisiologia , Carcinogênese/patologia , Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica/patologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/classificação , Tolerância a Radiação
6.
Anal Bioanal Chem ; 407(28): 8433-43, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26427501

RESUMO

Despite effective treatments, relapse of colorectal cancer (CRC) is frequent, in part caused by the existence of tumor-initiating cells (TICs). Different subtypes of TICs, quiescent and activated, coexist in tumors, defining the tumor aggressiveness and therapeutic response. These subtypes have been sorted by hyperlayer sedimentation field-flow fractionation (SdFFF) from WiDr and HCT116 cell lines. On the basis of a new strategy, including TIC SdFFF sorting, 3D Matrigel amplification, and grafting of corresponding TIC colonies on the chick chorioallantoic membrane (CAM), specific tumor matrices could be obtained. If tumors had similar architectural structure with vascularization by the host system, they had different proliferative indices in agreement with their initial quiescent or activated state. Protein analysis also revealed that tumors obtained from a population enriched for "activated" TICs lost "stemness" properties and became invasive. In contrast, tumors obtained from a population enriched for "quiescent" TICs kept their stemness properties and seemed to be less proliferative and invasive. Then, it was possible to produce different kinds of tumor which could be used as selective supports to study carcinogenesis and therapy sensitivity.


Assuntos
Biomarcadores Tumorais/genética , Separação Celular/métodos , Neoplasias Colorretais/diagnóstico , Modelos Biológicos , Células-Tronco Neoplásicas/classificação , Animais , Biomarcadores Tumorais/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Separação Celular/instrumentação , Embrião de Galinha , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/patologia , Colágeno/química , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Combinação de Medicamentos , Fracionamento por Campo e Fluxo/instrumentação , Fracionamento por Campo e Fluxo/métodos , Expressão Gênica , Células HCT116 , Humanos , Queratina-20/genética , Queratina-20/metabolismo , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Laminina/química , Invasividade Neoplásica , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/patologia , Proteoglicanas/química
7.
Ann Hematol ; 93(8): 1263-77, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24870942

RESUMO

Diffuse large B cell lymphoma (DLBCL) comprises specific subtypes, disease entities, and other not otherwise specified (NOS) lymphomas. This review will focus on DLBCL NOS because of their prevalence and their heterogeneity with respect to morphology, clinical presentation, biology, and response to treatment. Gene expression profiling of DLBCL NOS has identified molecular subgroups that correlate with prognosis and may have relevance for treatment based on signaling pathways. New technologies have revealed that the "activated B cell" subgroup is linked to activation of the nuclear factor kB (NF-kB) pathway, with mutations found in CD79A/B, CARD11, and MYD88, and loss of function mutations in TNFAIP3. The "germinal center B cell-like" subgroup is linked to mutational changes in EZH2 and CREBBP. Biomarkers that are related to pathways promoting tumor cell growth and survival in DLBCL have been recognized, although their predictive role requires clinical validation. Immunohistochemistry for detecting the expression of these biomarkers is a practical technique that could provide a rational for clinical trial design.


Assuntos
Biomarcadores Tumorais/análise , Linfoma Difuso de Grandes Células B/classificação , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Biomarcadores Tumorais/genética , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Linfoma Difuso de Grandes Células B/química , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/patologia , Terapia de Alvo Molecular , NF-kappa B/metabolismo , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/patologia , Terapias em Estudo , Microambiente Tumoral , Infecções Tumorais por Vírus
8.
Semin Cancer Biol ; 22(3): 250-60, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22459768

RESUMO

Despite advances in chemotherapy, hormone therapy and radiotherapy, not all cancer patients respond favorably to treatment. However, progress in understanding the mechanisms of malignant diseases and the mode of action of therapies are opening opportunities to match treatment to specific patient subpopulations, paving the way for personalized medicine. In this context, high throughput technologies that have been developed to determine gene expression profiles potentially offer an effective tool for dissecting the biology of cancer pathologies, for identifying candidate molecules for the development of new drugs, and for identifying individual patients who are more likely to respond favorably to a given therapy. Here, we overview and discuss the robustness of the deployment of these technologies in these contexts. We conclude that while these technologies are useful for target identification, there are limitations to their use in understanding cancer biology and in routine clinical application.


Assuntos
Perfilação da Expressão Gênica , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Humanos , Neoplasias/classificação , Neoplasias/diagnóstico , Células-Tronco Neoplásicas/classificação , Medicina de Precisão
9.
Stem Cells ; 30(1): 89-93, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22045578

RESUMO

Evidence gathered over the past two decades confirms earlier reports that suggested that hematologic malignancies exhibit a hierarchical differentiation structure similar to normal hematopoiesis. There is growing evidence that some solid tumors may also exhibit a differentiation program similar to the normal tissue of origin. Many excellent reviews on the topic of cancer stem cells (CSCs) document the recent explosion of information in the field, particularly highlighting the phenotypic and functional characteristics of these putative cells in vitro. Accordingly, here we only briefly discuss these concepts, and instead primarily examine the potential clinical relevance of CSCs, arguably the major unresolved issue in the field. Although it is generally accepted that CSCs are resistant to chemotherapy in vitro, only recently have data surfaced that suggest a role for these cells in disease relapse. Importantly, cancer cells with a stem cell phenotype have been found to be enriched in minimal residual disease of several malignancies. If the role of CSCs in relapse is confirmed, targeting these cells would hold substantial potential for improving the outcome of cancer patients.


Assuntos
Neoplasia Residual/etiologia , Células-Tronco Neoplásicas/fisiologia , Animais , Diferenciação Celular/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Leucemia/patologia , Camundongos , Neoplasias/patologia , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/citologia , Transdução de Sinais/genética
10.
Pancreatology ; 13(2): 110-3, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23561967

RESUMO

It is now well established that human pancreatic ductal adenocarcinoma (PDAC) contains a subset of cells with self-renewal capabilities and subsequent exclusive in vivo tumorigenic capacity as assessed by limiting dilution tumorigenic transplantation assays into immunodeficient mice. These cells are considered pancreatic cancer stem cells (CSCs) and are able to form tumors indistinguishable from parental ones. Furthermore they display strong chemotherapy resistance and are implicated in tumor relapses and metastatic spread. Important next steps for advancing the field of pancreatic CSC research include the identification and characterization of CSCs in the unperturbed in vivo setting. This has been achieved just recently for other solid tumors such as glioblastoma using clonal analysis after lineage tracing in mice [1]. In vivo imaging of CSCs during tumor development should not only provide new insights into the in vivo features of CSCs, but also help to further unravel the influence of the stroma on CSC biology. Comprehensive studies of the tumor heterogeneity with respect to the coexistence of different clones potentially generated by distinct population of CSCs that are evolving by stochastic cell fate decisions may actually unite the CSC concept and the model of clonal evolution for pancreatic cancer. Eventually, the design of specific therapies against CSCs should open new alleys to improve survival of patients with PDAC. Combined therapies targeting CSCs and their progenies as well as the supportive stroma may represent the most promising approach for the future treatment of patients with PDAC.


Assuntos
Adenocarcinoma/patologia , Células-Tronco Neoplásicas/citologia , Pâncreas/citologia , Neoplasias Pancreáticas/patologia , Adenocarcinoma/tratamento farmacológico , Animais , Antígenos CD/metabolismo , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Biomarcadores , Humanos , Camundongos , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico
11.
Lancet Oncol ; 13(1): e43-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22225725

RESUMO

Intratumoral heterogeneity in breast cancer is well documented. Although the mechanisms leading to this heterogeneity are not understood, a subpopulation of cancer cells, cancer stem cells (CSCs), that have some phenotypic similarities with adult tissue stem cells, has been suggested to contribute to tumour heterogeneity. It has been postulated that these CSCs are dormant, and by virtue of their low proliferative activity and ability to exclude intracellular toxins, are resistant to chemotherapy and radiation therapy. These cells were initially isolated based on the presence of markers such as CD44, CD24, and ALDH1, with further characterisation using mammosphere assay and transplantation into immunodeficient mice. The CSC hypothesis raises several theoretical and practical questions. Does cancer arise in normal mammary stem cells or do some malignant cells acquire a CSC phenotype through clonal evolution? Are CSCs in different molecular (intrinsic) subtypes of breast cancer similar, or do they have distinct properties based on the subtype? Does the CSC phenotype reflect plasticity or the dynamic nature of a few cancer cells? How do these cells acquire invasive behaviour, as they go through epithelial-to-mesenchymal transition and then revert to epithelial phenotype at sites of metastasis in response to tumour microenvironmental and metastasis site-specific cues? It is increasingly recognised that the methods and assays used for identifying CSCs have substantial limitations; does this negate the entire concept? In this Personal View, we argue that the CSC phenotype represents an aggressive clone that survives in an adverse environment through constant evolution and integration of various hallmarks of cancer. This evolution could involve acquiring mutations that permit asymmetric and symmetric division, converting the host immune attack to its own advantage, and plasticity to adapt to sites of metastasis through reversible change in adhesion molecules. We also argue that the cell-type origin of cancer could affect the rate at which CSCs develop in a tumour, with an eventual effect on disease outcome.


Assuntos
Neoplasias da Mama/patologia , Células-Tronco Neoplásicas/patologia , Semântica , Animais , Biomarcadores/metabolismo , Neoplasias da Mama/metabolismo , Comunicação Celular , Linhagem da Célula , Sobrevivência Celular , Transição Epitelial-Mesenquimal , Feminino , Humanos , Invasividade Neoplásica , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Prognóstico , Células Estromais/patologia , Terminologia como Assunto , Microambiente Tumoral
12.
Science ; 376(6596): eabe1505, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35617398

RESUMO

In castration-resistant prostate cancer (CRPC), the loss of androgen receptor (AR) dependence leads to clinically aggressive tumors with few therapeutic options. We used ATAC-seq (assay for transposase-accessible chromatin sequencing), RNA-seq, and DNA sequencing to investigate 22 organoids, six patient-derived xenografts, and 12 cell lines. We identified the well-characterized AR-dependent and neuroendocrine subtypes, as well as two AR-negative/low groups: a Wnt-dependent subtype, and a stem cell-like (SCL) subtype driven by activator protein-1 (AP-1) transcription factors. We used transcriptomic signatures to classify 366 patients, which showed that SCL is the second most common subtype of CRPC after AR-dependent. Our data suggest that AP-1 interacts with the YAP/TAZ and TEAD proteins to maintain subtype-specific chromatin accessibility and transcriptomic landscapes in this group. Together, this molecular classification reveals drug targets and can potentially guide therapeutic decisions.


Assuntos
Cromatina , Terapia de Alvo Molecular , Neoplasias de Próstata Resistentes à Castração , Linhagem Celular Tumoral , Cromatina/genética , Perfilação da Expressão Gênica , Humanos , Masculino , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo , Organoides/metabolismo , Organoides/patologia , Neoplasias de Próstata Resistentes à Castração/classificação , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
13.
Curr Med Sci ; 41(1): 127-132, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33582916

RESUMO

The stemness of different side population (SP) cell subtypes in ovarian cancer cells was studied, and the heterogeneity of ovarian cancer stem cells was analyzed. The cisplatin-resistant human serous ovarian cancer cell line C13 was stained with the bisbenzimide Hoechst 33342. A flow cytometry-based fluorescence-activated sorting method was used to obtain lower-SP (LSP) cells, upper-SP (USP) cells, and non-SP cells (NSP) based on their sensitivity to the staining time and Hoechst dye concentration. The sphere-forming capability, expression levels of stem cell markers, resistance to high concentrations of cisplatin, and subcutaneous tumorigenicity in NOD/SCID mice of the different cell subtypes were evaluated. The C13 cells contained SP cells with stemness characteristics, and the LSP cell subtype expressed higher levels of stem cell markers, had higher in vitro sphere-forming capability, higher cisplatin resistance and higher in vivo subcutaneous tumorigenesis than USP cells (P<0.05). NSP cells had no stemness. In conclusion, different subtypes of ovarian cancer SP cells have different stemness levels, and ovarian cancer stem cells may be heterogeneous.


Assuntos
Autorrenovação Celular , Células-Tronco Neoplásicas/classificação , Neoplasias Ovarianas/patologia , Animais , Antineoplásicos/farmacologia , Carcinogênese/patologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/fisiologia , Ensaio Tumoral de Célula-Tronco
14.
Genes (Basel) ; 12(11)2021 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-34828292

RESUMO

This study aims to investigate the differentiation trajectory of osteosarcoma cells and to construct molecular subtypes with their respective characteristics and generate a multi-gene signature for predicting prognosis. Integrated single-cell RNA-sequencing (scRNA-seq) data, bulk RNA-seq data and microarray data from osteosarcoma samples were used for analysis. Via scRNA-seq data, time-related as well as differentiation-related genes were recognized as osteosarcoma tumor stem cell-related genes (OSCGs). In Gene Expression Omnibus (GEO) cohort, osteosarcoma patients were classified into two subtypes based on prognostic OSCGs and it was found that molecular typing successfully predicted overall survival, tumor microenvironment and immune infiltration status. Further, available drugs for influencing osteosarcoma via prognostic OSCGs were revealed. A 3-OSCG-based prognostic risk score signature was generated and by combining other clinic-pathological independent prognostic factor, stage at diagnosis, a nomogram was established to predict individual survival probability. In external independent TARGET cohort, the molecular types, the 3-gene signature as well as nomogram were validated. In conclusion, osteosarcoma cell differentiation occupies a crucial position in many facets, such as tumor prognosis and microenvironment, suggesting promising therapeutic targets for this disease.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Ósseas/classificação , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Osteossarcoma/classificação , Neoplasias Ósseas/genética , Neoplasias Ósseas/mortalidade , Bases de Dados Genéticas , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/classificação , Análise de Sequência com Séries de Oligonucleotídeos , Osteossarcoma/genética , Osteossarcoma/mortalidade , Prognóstico , RNA-Seq , Análise de Célula Única , Análise de Sobrevida , Microambiente Tumoral
15.
STAR Protoc ; 2(2): 100484, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33982017

RESUMO

BMI1-expressing cancer stem cells (CSCs) play a key role in the development, progression, therapy resistance, recurrence, and metastasis of head and neck squamous cell carcinoma (HNSCC). Here, we present a chemically-induced HNSCC mouse model, genetically and pathologically similar to human HNSCC. This protocol describes how to use genetic lineage tracing based on the Cre-loxP recombination strategy, which allows us to study the regulation and targeting of BMI1+ CSCs in primary tumors and lymph node metastases. For complete details on the use and execution of this protocol, please refer to Chen et al. (2017) and Jia et al. (2020).


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias Experimentais , Células-Tronco Neoplásicas , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Linhagem da Célula/genética , Células Cultivadas , Feminino , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Imuno-Histoquímica , Masculino , Camundongos , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 1/química , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia
16.
Cell Mol Biol (Noisy-le-grand) ; 56 Suppl: OL1350-8, 2010 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-20937222

RESUMO

We studied the tumor stem cell properties of the CD133+CD44+ subpopulation in the human lung adenocarcinoma cell line A549. A549 cells were classified into subpopulations based on differential expression patterns for CD133 and CD44. Cells from different subpopulations were cultured and subcutaneously injected into 32 nude mice. Our results as following, (1) The majority of A549 cells died, whereas only about 4.11% of cells divided and proliferated to form cell clones. (2) The expression of CD133 and CD44 in proliferative cancer cells was statistically significantly different from that in normal A549 cells (p < 0.001). (3) Cell proliferation in group A (CD133+CD44+) was the fastest among all groups. Cell proliferation in A549 cells was slower than in group A but faster than in groups B (CD133-CD44-), C (CD133-CD44+), and D (CD133+CD44-). (4) The tumorigenic capacity in cells from group A was significantly higher than that in cells from groups B (p<0.001), C (p<0.001) and D (p<0.04). In conclusion, CD133+CD44+ cells in the adenocarcinoma cell line A549 have expressive significant cancer stem cell properties with continuous proliferative capacity and differentiation potential.


Assuntos
Adenocarcinoma/patologia , Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Receptores de Hialuronatos/metabolismo , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/patologia , Peptídeos/metabolismo , Antígeno AC133 , Adenocarcinoma/classificação , Adenocarcinoma/metabolismo , Animais , Antígenos CD/genética , Testes de Carcinogenicidade , Linhagem Celular Tumoral , Proliferação de Células , Imunofluorescência , Glicoproteínas/genética , Humanos , Receptores de Hialuronatos/genética , Injeções Subcutâneas , Neoplasias Pulmonares/classificação , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo , Peptídeos/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo
17.
Methods Mol Biol ; 2129: 177-191, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32056178

RESUMO

Cancer stem cells (CSCs) are a small subpopulation of cells associated with cancer initiation, progression, metastasis, therapy resistant, and recurrence. In esophageal squamous cell carcinoma (ESCC), several cell surface and intracellular markers, for example, CD44, ALDH, Pygo2, MAML1, Twist1, Musashi1, side population (SP), CD271, and CD90, have been proposed to identify CSCs. In addition, stem cell markers such as ALDH1, HIWI, Oct3/4, ABCG2, SOX2, SALL4, BMI-1, NANOG, CD133, and podoplanin were associated with pathological stages of cancer, cancer recurrence, prognosis, and therapy resistance of patients with ESCC. Identification and isolation of CSCs could play an important part of improved cancer management regime in ESCC. Furthermore, CSCs may be used as the predictive tool for chemoradiotherapy response in ESCC. Different methods such as in vitro functional assays, cell sorting using various intracellular, and cell surface markers and xenotransplantation techniques are frequently used for the identification and isolation of CSCs in different cancers, including ESCC. However, none of these methods solely can guarantee complete isolation of CSC population. Therefore, a combination of methods is used for reliable detection and isolation of CSCs. Herein, we describe the identification and isolation of CSCs from ESCC cells by cell sorting after Hoechst 33342 staining followed by in vitro functional assays and in vivo mouse xenotransplantation techniques.


Assuntos
Carcinoma de Células Escamosas do Esôfago/patologia , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/patologia , Animais , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Humanos , Masculino , Camundongos , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/metabolismo , Prognóstico
18.
J Hematol Oncol ; 13(1): 63, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32493379

RESUMO

Here, we report a unique acute myeloid leukemia (AML) bone marrow-derived mesenchymal stem cell (MSC) with both mesenchymal and endothelial potential, which we have named Mesenchymal Cancer Stem Cells (MCSCs). These MCSCs are CD90-CD13-CD44+ and differ from MSCs in isolation, expansion, differentiation, immunophenotype, and cytokine release profile. Furthermore, blocking CD44 inhibited the proliferation and cluster formation of early MCSCs with lower ICAM-1 protein levels. Similar CD90-CD44+ cancer stem cells have been reported in both gastric and breast cancers, which grew in floating spheres in vitro and exhibited mesenchymal features and high metastatic/tumorigenic capabilities in vivo. Our novel discovery provides the first evidence that certain AMLs may be comprised of both hematopoietic and stromal malignant cells. Targeting MCSCs and their cytokine release has potential as a novel therapeutic approach in AML.


Assuntos
Antígenos de Neoplasias/análise , Medula Óssea/patologia , Receptores de Hialuronatos/análise , Leucemia Mielomonocítica Aguda/patologia , Células-Tronco Mesenquimais/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Angiogênicas/metabolismo , Adesão Celular , Separação Celular , Citocinas/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Leucemia Mielomonocítica Aguda/metabolismo , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/metabolismo , Células Tumorais Cultivadas
19.
Nat Commun ; 11(1): 991, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080185

RESUMO

Characterizing the complex composition of solid tumors is fundamental for understanding tumor initiation, progression and metastasis. While patient-derived samples provide valuable insight, they are heterogeneous on multiple molecular levels, and often originate from advanced tumor stages. Here, we use single-cell transcriptome and epitope profiling together with pathway and lineage analyses to study tumorigenesis from a developmental perspective in a mouse model of salivary gland squamous cell carcinoma. We provide a comprehensive cell atlas and characterize tumor-specific cells. We find that these cells are connected along a reproducible developmental trajectory: initiated in basal cells exhibiting an epithelial-to-mesenchymal transition signature, tumorigenesis proceeds through Wnt-differential cancer stem cell-like subpopulations before differentiating into luminal-like cells. Our work provides unbiased insights into tumor-specific cellular identities in a whole tissue environment, and emphasizes the power of using defined genetic model systems.


Assuntos
Carcinogênese/genética , Carcinogênese/patologia , Animais , Carcinogênese/imunologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/imunologia , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neoplásicas/classificação , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , RNA Mensageiro/genética , RNA-Seq , Neoplasias das Glândulas Salivares/genética , Neoplasias das Glândulas Salivares/imunologia , Neoplasias das Glândulas Salivares/patologia , Análise de Célula Única , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
20.
Theranostics ; 10(7): 3083-3098, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194856

RESUMO

Over a decade of research has confirmed the critical role of cancer stem-like cells (CSCs) in tumor initiation, chemoresistance, and metastasis. Increasingly, CSC hierarchies have begun to be defined with some recurring themes. This includes evidence that these hierarchies are 'flexible,' with both cell state transitions and dedifferentiation events possible. These findings pose therapeutic hurdles and opportunities. Here, we review cancer stem cell hierarchies and their interactions with the tumor microenvironment. We also discuss the current therapeutic approaches designed to target CSC hierarchies and initial clinical trial results for CSC targeting agents. While cancer stem cell targeted therapies are still in their infancy, we are beginning to see encouraging results that suggest a positive outlook for CSC-targeting approaches.


Assuntos
Células-Tronco Neoplásicas/classificação , Família Aldeído Desidrogenase 1/antagonistas & inibidores , Animais , Antígenos CD/efeitos dos fármacos , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais , Neoplasias Encefálicas/patologia , Neoplasias da Mama/patologia , Desdiferenciação Celular , Linhagem da Célula , Ensaios Clínicos como Assunto , Epigênese Genética , Feminino , Humanos , Metformina/farmacologia , Metformina/uso terapêutico , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA