Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cancer Ther ; 22(12): 1444-1453, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37619980

RESUMO

Integrin beta-6, a component of the heterodimeric adhesion receptor alpha-v/beta-6, is overexpressed in numerous solid tumors. Its expression has been shown by multiple investigators to be a negative prognostic indicator in diverse cancers including colorectal, non-small cell lung, gastric, and cervical. We developed SGN-B6A as an antibody-drug conjugate (ADC) directed to integrin beta-6 to deliver the clinically validated payload monomethyl auristatin E (MMAE) to cancer cells. The antibody component of SGN-B6A is specific for integrin beta-6 and does not bind other alpha-v family members. In preclinical studies, this ADC has demonstrated activity in vivo in models derived from non-small cell lung, pancreatic, pharyngeal, and bladder carcinomas spanning a range of antigen expression levels. In nonclinical toxicology studies in cynomolgus monkeys, doses of up to 5 mg/kg weekly for four doses or 6 mg/kg every 3 weeks for two doses were tolerated. Hematologic toxicities typical of MMAE ADCs were dose limiting, and no significant target-mediated toxicity was observed. A phase I first-in-human study is in progress to evaluate the safety and antitumor activity of SGN-B6A in a variety of solid tumors known to express integrin beta-6 (NCT04389632).


Assuntos
Antineoplásicos , Carcinoma , Imunoconjugados , Humanos , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Prognóstico , Integrinas , Linhagem Celular Tumoral
2.
JCO Precis Oncol ; 6: e2100280, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35294224

RESUMO

PURPOSE: Patients with metastatic triple-negative breast cancer (mTNBC) have poor outcomes. The Intensive Trial of Omics in Cancer (ITOMIC) sought to determine the feasibility and potential efficacy of informing treatment decisions through multiple biopsies of mTNBC deposits longitudinally over time, accompanied by analysis using a distributed network of experts. METHODS: Thirty-one subjects were enrolled and 432 postenrollment biopsies performed (clinical and study-directed) of which 332 were study-directed. Molecular profiling included whole-genome sequencing or whole-exome sequencing, cancer-associated gene panel sequencing, RNA-sequencing, and immunohistochemistry. To afford time for analysis, subjects were initially treated with cisplatin (19 subjects), or another treatment they had not received previously. The results were discussed at a multi-institutional ITOMIC Tumor Board, and a report transmitted to the subject's oncologist who arrived at the final treatment decision in conjunction with the subject. Assistance was provided to access treatments that were predicted to be effective. RESULTS: Multiple biopsies in single settings and over time were safe, and comprehensive analysis was feasible. Two subjects were found to have lung cancer, one had carcinoma of unknown primary site, tumor samples from three subjects were estrogen receptor-positive and from two others, human epidermal growth factor receptor 2-positive. Two subjects withdrew. Thirty-four of 112 recommended treatments were accessed using approved drugs, clinical trials, and single-patient investigational new drugs. After excluding the three subjects with nonbreast cancers and the two subjects who withdrew, 22 of 26 subjects (84.6%) received at least one ITOMIC Tumor Board-recommended treatment. CONCLUSION: Further exploration of this approach in patients with mTNBC is merited.


Assuntos
Neoplasias de Mama Triplo Negativas , Cisplatino/uso terapêutico , Estudos de Viabilidade , Humanos , Técnicas de Diagnóstico Molecular , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
3.
BMC Cancer ; 19(1): 220, 2019 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-30871481

RESUMO

BACKGROUND: Breast cancer patient-derived xenograft (BC-PDX) models represent a continuous and reproducible source of circulating tumor cells (CTCs) for studying their role in tumor biology and metastasis. We have previously shown the utility of BC-PDX models in the study of CTCs by immunohistochemistry (IHC) on serial paraffin sections and manual microscopic identification of cytokeratin-positive cells, a method that is both low-throughput and labor-intensive. We therefore aimed to identify and characterize CTCs from small volume mouse blood samples and examined its practical workflow in a study of BC-PDX mice treated with chemotherapy using an automated imaging platform, the AccuCyte®-CyteFinder® system. METHODS: CTC analysis was conducted using blood from non-tumor bearing SCID/Beige mice spiked with human breast cancer cells, BC-PDX-bearing mice, and BC-PDX mice treated with vehicle or chemotherapeutic agent(s). After red blood cell lysis, nucleated cells were mixed with transfer solution, processed onto microscope slides, and stained by immunofluorescence. The CyteFinder automated scanning microscope was used to identify CTCs, defined as nucleated cells that were human cytokeratin-positive, and mouse CD45-negative. Disaggregated primary BC-PDX tumors and lung metastatic nodules were processed using the same immunostaining protocol. Collective expression of breast cancer cell surface markers (EpCAM, EGFR, and HER2) using a cocktail of target-specific antibodies was assessed. CTCs and disaggregated tumor cells were individually retrieved from slides using the CytePicker® module for sequence analysis of a BC-PDX tumor-specific PIK3CA mutation. RESULTS: The recovery rate of human cancer cells spiked into murine blood was 83 ± 12%. CTC detection was not significantly different from the IHC method. One-third of CTCs did not stain positive for cell surface markers. A PIK3CA T1035A mutation present in a BC-PDX tumor was confirmed in isolated single CTCs and cells from dissociated metastatic nodules after whole genome amplification and sequencing. CTC evaluation could be simply implemented into a preclinical PDX therapeutic study setting with substantial improvements in workflow over the IHC method. CONCLUSIONS: Analysis of small volume blood samples from BC-PDX-bearing mice using the AccuCyte-CyteFinder system allows investigation of the role of CTCs in tumor biology and metastasis independent of surface marker expression.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/genética , Células Neoplásicas Circulantes/metabolismo , Análise de Célula Única/métodos , Animais , Antineoplásicos/farmacologia , Biomarcadores Tumorais/sangue , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Separação Celular , Classe I de Fosfatidilinositol 3-Quinases/sangue , Feminino , Humanos , Queratinas/sangue , Antígenos Comuns de Leucócito/sangue , Camundongos , Camundongos SCID , Mutação , Transplante de Neoplasias , Células Neoplásicas Circulantes/efeitos dos fármacos , Análise de Sequência de DNA
4.
Prenat Diagn ; 38(13): 1069-1078, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30357877

RESUMO

OBJECTIVE: To gather additional data on the ability to detect subchromosomal abnormalities of various sizes in single fetal cells isolated from maternal blood, using low-coverage shotgun next-generation sequencing for cell-based noninvasive prenatal testing (NIPT). METHOD: Fetal trophoblasts were recovered from approximately 30 mL of maternal blood using maternal white blood cell depletion, density-based cell separation, immunofluorescence staining, and high-resolution scanning. These trophoblastic cells were picked as single cells and underwent whole genome amplification for subsequent genome-wide copy number analysis and genotyping to confirm the fetal origin of the cells. RESULTS: Applying our fetal cell isolation method to a series of 125 maternal blood samples, we detected on average 4.17 putative fetal cells/sample. The series included 15 cases with clinically diagnosed fetal aneuploidies and five cases with subchromosomal abnormalities. This method was capable of detecting findings that were 1 to 2 Mb in size, and all were concordant with the microarray or karyotype data obtained on a fetal sample. A minority of fetal cells showed evidence of genome degradation likely related to apoptosis. CONCLUSION: We demonstrate that this cell-based NIPT method has the capacity to reliably diagnose fetal chromosomal abnormalities down to 1 to 2 Mb in size.


Assuntos
Aberrações Cromossômicas , Variações do Número de Cópias de DNA , Deleção de Genes , Duplicação Gênica , Trofoblastos/metabolismo , Adulto , Separação Celular , Deleção Cromossômica , Feminino , Imunofluorescência , Genótipo , Humanos , Cariótipo , Masculino , Gravidez , Diagnóstico Pré-Natal , Análise de Célula Única , Análise Serial de Tecidos , Adulto Jovem
5.
Methods Mol Biol ; 1634: 163-172, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819849

RESUMO

The RareCyte platform addresses important technology limitations of current circulating tumor cell (CTC) collection methods, and expands CTC interrogation to include advanced phenotypic characterization and single-cell molecular analysis. In this respect, it represents the "next generation" of cell-based liquid biopsy technologies. In order to identify and analyze CTCs, RareCyte has developed an integrated sample preparation, imaging and individual cell retrieval process. The first step in the process, AccuCyte®, allows the separation, collection, and transfer to a slide the nucleated cell fraction of the blood that contains CTCs. Separation and collection are based on cell density-rather than size or surface molecular expression-and are performed within a closed system, without wash or lysis steps, enabling high CTC recovery. Here, we describe our technique for nucleated cell collection from a blood sample, and the spreading of these nucleated cells onto glass slides permitting immunofluorescent staining, cell identification, and individual cell picking described in subsequent chapters. In addition to collection of rare cells such as CTCs, AccuCyte also collects cells of the circulating immune system onto archivable slides as well as plasma from the same sample.


Assuntos
Separação Celular/métodos , Células Imobilizadas/patologia , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patologia , Análise de Célula Única/métodos , Contagem de Células , Linhagem Celular Tumoral , Separação Celular/instrumentação , Células Imobilizadas/metabolismo , Centrifugação/instrumentação , Centrifugação/métodos , Desenho de Equipamento , Humanos , Neoplasias/sangue , Neoplasias/patologia , Células Neoplásicas Circulantes/metabolismo , Kit de Reagentes para Diagnóstico/normas , Análise de Célula Única/instrumentação
6.
Methods Mol Biol ; 1634: 173-180, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819850

RESUMO

The RareCyte CyteFinder instrument is an automated scanner that allows rapid identification of circulating tumor cells (CTCs) on microscope slides prepared by the AccuCyte process (see Chapter 13 ) and stained by immunofluorescence. Here, we present the workflow for CyteFinder scanning, analysis, and CyteMapper scan review which includes CTC confirmation and report generation.


Assuntos
Separação Celular/métodos , Células Imobilizadas/patologia , Processamento de Imagem Assistida por Computador/métodos , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patologia , Análise de Célula Única/métodos , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Automação Laboratorial/instrumentação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Contagem de Células , Linhagem Celular Tumoral , Separação Celular/instrumentação , Células Imobilizadas/imunologia , Células Imobilizadas/metabolismo , Centrifugação/instrumentação , Centrifugação/métodos , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Desenho de Equipamento , Imunofluorescência/métodos , Corantes Fluorescentes/química , Humanos , Imunoconjugados/química , Queratinas/genética , Queratinas/imunologia , Queratinas/metabolismo , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Antígenos Comuns de Leucócito/metabolismo , Neoplasias/sangue , Neoplasias/imunologia , Neoplasias/patologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Ligação Proteica , Análise de Célula Única/instrumentação
7.
Methods Mol Biol ; 1634: 181-192, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819851

RESUMO

The CytePicker module built into the RareCyte CyteFinder instrument allows researchers to easily retrieve individual cells from microscope slides for genomic analyses, including array CGH, targeted sequencing, and next-generation sequencing. Here, we describe the semiautomated retrieval of CTCs from the blood processed by AccuCyte (see Chapter 13) and amplification of genomic DNA so that molecular analysis can be performed.


Assuntos
Separação Celular/métodos , Células Imobilizadas/patologia , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patologia , Técnicas de Amplificação de Ácido Nucleico/métodos , Análise de Célula Única/métodos , Automação Laboratorial/instrumentação , Contagem de Células , Linhagem Celular Tumoral , Separação Celular/instrumentação , Células Imobilizadas/imunologia , Células Imobilizadas/metabolismo , Centrifugação/instrumentação , Centrifugação/métodos , Hibridização Genômica Comparativa , Desenho de Equipamento , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/sangue , Neoplasias/imunologia , Neoplasias/patologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/metabolismo , Análise de Célula Única/instrumentação
8.
Prenat Diagn ; 36(11): 1009-1019, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27616633

RESUMO

OBJECTIVE: The goal was to develop methods for detection of chromosomal and subchromosomal abnormalities in fetal cells in the mother's circulation at 10-16 weeks' gestation using analysis by array comparative genomic hybridization (CGH) and/or next-generation sequencing (NGS). METHOD: Nucleated cells from 30 mL of blood collected at 10-16 weeks' gestation were separated from red cells by density fractionation and then immunostained to identify cytokeratin positive and CD45 negative trophoblasts. Individual cells were picked and subjected to whole genome amplification, genotyping, and analysis by array CGH and NGS. RESULTS: Fetal cells were recovered from most samples as documented by Y chromosome PCR, short tandem repeat analysis, array CGH, and NGS including over 30 normal male cells, one 47,XXY cell from an affected fetus, one trisomy 18 cell from an affected fetus, nine cells from a trisomy 21 case, three normal cells and one trisomy 13 cell from a case with confined placental mosaicism, and two chromosome 15 deletion cells from a case known by CVS to have a 2.7 Mb de novo deletion. CONCLUSION: We believe that this is the first report of using array CGH and NGS whole genome sequencing to detect chromosomal abnormalities in fetal trophoblastic cells from maternal blood. © 2016 The Authors. Prenatal Diagnosis published by John Wiley & Sons, Ltd.


Assuntos
Aberrações Cromossômicas , Hibridização Genômica Comparativa , Testes para Triagem do Soro Materno/métodos , Análise de Sequência de DNA , Trofoblastos/citologia , Variações do Número de Cópias de DNA , Estudos de Viabilidade , Feminino , Voluntários Saudáveis , Humanos , Masculino , Gravidez
9.
J Natl Compr Canc Netw ; 14(1): 8-17, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26733551

RESUMO

Accelerating cancer research is expected to require new types of clinical trials. This report describes the Intensive Trial of OMics in Cancer (ITOMIC) and a participant with triple-negative breast cancer metastatic to bone, who had markedly elevated circulating tumor cells (CTCs) that were monitored 48 times over 9 months. A total of 32 researchers from 14 institutions were engaged in the patient's evaluation; 20 researchers had no prior involvement in patient care and 18 were recruited specifically for this patient. Whole-exome sequencing of 3 bone marrow samples demonstrated a novel ROS1 variant that was estimated to be present in most or all tumor cells. After an initial response to cisplatin, a hypothesis of crizotinib sensitivity was disproven. Leukapheresis followed by partial CTC enrichment allowed for the development of a differential high-throughput drug screen and demonstrated sensitivity to investigational BH3-mimetic inhibitors of BCL-2 that could not be tested in the patient because requests to the pharmaceutical sponsors were denied. The number and size of CTC clusters correlated with clinical status and eventually death. Focusing the expertise of a distributed network of investigators on an intensively monitored patient with cancer can generate high-resolution views of the natural history of cancer and suggest new opportunities for therapy. Optimization requires access to investigational drugs.


Assuntos
Redes Comunitárias , Pesquisadores , Neoplasias de Mama Triplo Negativas/diagnóstico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/secundário , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Prova Pericial , Feminino , Seguimentos , Humanos , Leucaférese , Estudos Longitudinais , Pessoa de Meia-Idade , Metástase Neoplásica , Células Neoplásicas Circulantes , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/terapia
10.
BMC Cancer ; 15: 360, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25944336

RESUMO

BACKGROUND: Circulating tumor cells (CTCs) are malignant cells that have migrated from solid cancers into the blood, where they are typically present in rare numbers. There is great interest in using CTCs to monitor response to therapies, to identify clinically actionable biomarkers, and to provide a non-invasive window on the molecular state of a tumor. Here we characterize the performance of the AccuCyte®--CyteFinder® system, a comprehensive, reproducible and highly sensitive platform for collecting, identifying and retrieving individual CTCs from microscopic slides for molecular analysis after automated immunofluorescence staining for epithelial markers. METHODS: All experiments employed a density-based cell separation apparatus (AccuCyte) to separate nucleated cells from the blood and transfer them to microscopic slides. After staining, the slides were imaged using a digital scanning microscope (CyteFinder). Precisely counted model CTCs (mCTCs) from four cancer cell lines were spiked into whole blood to determine recovery rates. Individual mCTCs were removed from slides using a single-cell retrieval device (CytePicker™) for whole genome amplification and subsequent analysis by PCR and Sanger sequencing, whole exome sequencing, or array-based comparative genomic hybridization. Clinical CTCs were evaluated in blood samples from patients with different cancers in comparison with the CellSearch® system. RESULTS: AccuCyte--CyteFinder presented high-resolution images that allowed identification of mCTCs by morphologic and phenotypic features. Spike-in mCTC recoveries were between 90 and 91%. More than 80% of single-digit spike-in mCTCs were identified and even a single cell in 7.5 mL could be found. Analysis of single SKBR3 mCTCs identified presence of a known TP53 mutation by both PCR and whole exome sequencing, and confirmed the reported karyotype of this cell line. Patient sample CTC counts matched or exceeded CellSearch CTC counts in a small feasibility cohort. CONCLUSION: The AccuCyte--CyteFinder system is a comprehensive and sensitive platform for identification and characterization of CTCs that has been applied to the assessment of CTCs in cancer patient samples as well as the isolation of single cells for genomic analysis. It thus enables accurate non-invasive monitoring of CTCs and evolving cancer biology for personalized, molecularly-guided cancer treatment.


Assuntos
Separação Celular/métodos , Células Neoplásicas Circulantes , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Hibridização Genômica Comparativa , Análise Mutacional de DNA , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Masculino , Neoplasias da Próstata/patologia , Análise de Célula Única
11.
Clin Cancer Res ; 13(19): 5745-55, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908964

RESUMO

PURPOSE: This study was designed to elucidate the role of amplification at 8q24 in the pathophysiology of ovarian and breast cancer because increased copy number at this locus is one of the most frequent genomic abnormalities in these cancers. EXPERIMENTAL DESIGN: To accomplish this, we assessed the association of amplification at 8q24 with outcome in ovarian cancers using fluorescence in situ hybridization to tissue microarrays and measured responses of ovarian and breast cancer cell lines to specific small interfering RNAs against the oncogene MYC and a putative noncoding RNA, PVT1, both of which map to 8q24. RESULTS: Amplification of 8q24 was associated with significantly reduced survival duration. In addition, small interfering RNA-mediated reduction in either PVT1 or MYC expression inhibited proliferation in breast and ovarian cancer cell lines in which they were both amplified and overexpressed but not in lines in which they were not amplified/overexpressed. Inhibition of PVT1 expression also induced a strong apoptotic response in cell lines in which it was overexpressed but not in lines in which it was not amplified/overexpressed. Inhibition of MYC, on the other hand, did not induce an apoptotic response in cell lines in which MYC was amplified and overexpressed. CONCLUSIONS: These results suggest that MYC and PVT1 contribute independently to ovarian and breast pathogenesis when overexpressed because of genomic abnormalities. They also suggest that PVT1-mediated inhibition of apoptosis may explain why amplification of 8q24 is associated with reduced survival duration in patients treated with agents that act through apoptotic mechanisms.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Cromossomos Humanos Par 8 , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/fisiopatologia , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Apoptose , Neoplasias da Mama/mortalidade , Aberrações Cromossômicas , Feminino , Perfilação da Expressão Gênica , Genoma , Humanos , Hibridização in Situ Fluorescente , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Proteínas Proto-Oncogênicas c-myc/biossíntese , RNA Longo não Codificante , Transcrição Gênica , Resultado do Tratamento
12.
Methods Mol Biol ; 356: 353-65, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16988415

RESUMO

Cancers result from large-scale deregulation of genes that lead to cancer pathophysiologies such as increase proliferation, decreased apoptosis, increased motility, increased angiogenesis, and others. Genes that influence proliferation and apoptosis are particularly attractive as therapeutic targets. To identify genes that influence these phenotypes, we have developed simple and rapid methods to measure apoptosis and cell proliferation using high content screening with YO-PRO-1 and anti-BrdU staining of BrdU pulsed cells, respectively.


Assuntos
Genômica , Neoplasias Ovarianas/metabolismo , Biologia de Sistemas , Análise Serial de Tecidos , Benzimidazóis/metabolismo , Bromodesoxiuridina , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/metabolismo , Coloração e Rotulagem , Estatística como Assunto
13.
Cancer Cell ; 10(6): 515-27, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17157791

RESUMO

Recent studies suggest that thousands of genes may contribute to breast cancer pathophysiologies when deregulated by genomic or epigenomic events. Here, we describe a model "system" to appraise the functional contributions of these genes to breast cancer subsets. In general, the recurrent genomic and transcriptional characteristics of 51 breast cancer cell lines mirror those of 145 primary breast tumors, although some significant differences are documented. The cell lines that comprise the system also exhibit the substantial genomic, transcriptional, and biological heterogeneity found in primary tumors. We show, using Trastuzumab (Herceptin) monotherapy as an example, that the system can be used to identify molecular features that predict or indicate response to targeted therapies or other physiological perturbations.


Assuntos
Neoplasias da Mama/classificação , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genômica , Humanos , Proteínas de Neoplasias/análise
14.
Nano Lett ; 6(4): 800-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16608287

RESUMO

Quantum dots (Qdots) are now used extensively for labeling in biomedical research, and this use is predicted to grow because of their many advantages over alternative labeling methods. Uncoated Qdots made of core/shell CdSe/ZnS are toxic to cells because of the release of Cd2+ ions into the cellular environment. This problem has been partially overcome by coating Qdots with polymers, poly(ethylene glycol) (PEG), or other inert molecules. The most promising coating to date, for reducing toxicity, appears to be PEG. When PEG-coated silanized Qdots (PEG-silane-Qdots) are used to treat cells, toxicity is not observed, even at dosages above 10-20 nM, a concentration inducing death when cells are treated with polymer or mercaptoacid coated Qdots. Because of the importance of Qdots in current and future biomedical and clinical applications, we believe it is essential to more completely understand and verify this negative global response from cells treated with PEG-silane-Qdots. Consequently, we examined the molecular and cellular response of cells treated with two different dosages of PEG-silane-Qdots. Human fibroblasts were exposed to 8 and 80 nM of these Qdots, and both phenotypic as well as whole genome expression measurements were made. PEG-silane-Qdots did not induce any statistically significant cell cycle changes and minimal apoptosis/necrosis in lung fibroblasts (IMR-90) as measured by high content image analysis, regardless of the treatment dosage. A slight increase in apoptosis/necrosis was observed in treated human skin fibroblasts (HSF-42) at both the low and the high dosages. We performed genome-wide expression array analysis of HSF-42 exposed to doses 8 and 80 nM to link the global cell response to a molecular and genetic phenotype. We used a gene array containing approximately 22,000 total probe sets, containing 18,400 probe sets from known genes. Only approximately 50 genes (approximately 0.2% of all the genes tested) exhibited a statistically significant change in expression level of greater than 2-fold. Genes activated in treated cells included those involved in carbohydrate binding, intracellular vesicle formation, and cellular response to stress. Conversely, PEG-silane-Qdots induce a down-regulation of genes involved in controlling the M-phase progression of mitosis, spindle formation, and cytokinesis. Promoter analysis of these results reveals that expression changes may be attributed to the down-regulation of FOXM and BHLB2 transcription factors. Remarkably, PEG-silane-Qdots, unlike carbon nanotubes, do not activate genes indicative of a strong immune and inflammatory response or heavy-metal-related toxicity. The experimental evidence shows that CdSe/ZnS Qdots, if appropriately protected, induce negligible toxicity to the model cell system studied here, even when exposed to high dosages. This study indicates that PEG-coated silanized Qdots pose minimal impact to cells and are a very promising alternative to uncoated Qdots.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteoma/metabolismo , Pontos Quânticos , Dióxido de Silício/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Humanos
15.
Nano Lett ; 5(12): 2448-64, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16351195

RESUMO

The increasing use of nanotechnology in consumer products and medical applications underlies the importance of understanding its potential toxic effects to people and the environment. Although both fullerene and carbon nanotubes have been demonstrated to accumulate to cytotoxic levels within organs of various animal models and cell types and carbon nanomaterials have been exploited for cancer therapies, the molecular and cellular mechanisms for cytotoxicity of this class of nanomaterial are not yet fully apparent. To address this question, we have performed whole genome expression array analysis and high content image analysis based phenotypic measurements on human skin fibroblast cell populations exposed to multiwall carbon nano-onions (MWCNOs) and multiwall carbon nanotubes (MWCNTs). Here we demonstrate that exposing cells to MWCNOs and MWCNTs at cytotoxic doses induces cell cycle arrest and increases apoptosis/necrosis. Expression array analysis indicates that multiple cellular pathways are perturbed after exposure to these nanomaterials at these doses, with material-specific toxigenomic profiles observed. Moreover, there are also distinct qualitative and quantitative differences in gene expression profiles, with each material at different dosage levels (6 and 0.6 microg/mL for MWCNO and 0.6 and 0.06 microg/mL for MWCNT). MWCNO and MWCNT exposure activates genes involved in cellular transport, metabolism, cell cycle regulation, and stress response. MWCNTs induce genes indicative of a strong immune and inflammatory response within skin fibroblasts, while MWCNO changes are concentrated in genes induced in response to external stimuli. Promoter analysis of the microarray results demonstrate that interferon and p38/ERK-MAPK cascades are critical pathway components in the induced signal transduction contributing to the more adverse effects observed upon exposure to MWCNTs as compared to MWCNOs.


Assuntos
Fibroblastos/metabolismo , Fibroblastos/patologia , Nanotubos de Carbono/efeitos adversos , Proteoma/metabolismo , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Teste de Materiais , Nanotubos de Carbono/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia
16.
Mol Ther ; 9(3): 337-46, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15006600

RESUMO

The role of the virion shell in viral pathogenesis is relatively unknown yet the use of viral vectors in human gene transfer experiments requires an understanding of these interactions. In this study, we used DNA microarrays to identify genes modulated during pathogenic adenovirus or nonpathogenic adeno-associated virus infections. Responses to wt viruses, recombinant vectors, or empty virion particles were compared. Adeno-associated virus shells induced nearly the full complement of changes elicited by the intact virus. The cellular genes elicited a nonpathogenic response, with antiproliferative genes being induced as a cluster. In contrast, adenovirus and adenovirus empty capsid infection yielded a broader response and subset, respectively, including induction of immune and stress-response genes associated with pathogenic effects. Our studies show that the impact of the viral capsid on cellular gene expression, and potential host toxicity, must be considered independent of the vector genome for safe gene transfer in the clinic.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Vírion/genética , Vírus/genética , Adenoviridae/genética , Capsídeo/metabolismo , Ciclo Celular , Linhagem Celular , DNA/metabolismo , Dependovirus/genética , Feminino , Feto/metabolismo , Fibroblastos/metabolismo , Fase G2 , Terapia Genética/métodos , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Mitose , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo
17.
J Virol ; 77(23): 12881-5, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14610209

RESUMO

Adenovirus (Ad) has been extensively studied as a eukaryotic viral vector. As these vectors have evolved from first-generation vectors to vectors that contain either very few or no viral genes ("gutless" Ad), significant reductions in the host innate immune response upon infection have been observed. Regardless of these vector improvements an unknown amount of toxicity has been associated with the virion structural proteins. Here we demonstrate the ability to generate high particle numbers (10(11) to 10(12)) of Ad empty virions based on a modification of Cre/lox gutless Ad vectors. Using a battery of analyses (electron microscopy, atomic force microscopy, confocal images, and competition assays) we characterized this reagent and determined that it (i) makes intact virion particles, (ii) competes for receptor binding with wild-type Ad, and (iii) enters the cell proficiently, demonstrating an ability to carry out essential steps of viral entry. To further study the biological impact of these Ad empty virions on infected cells, we carried out DNA microarray analysis. Compared to that for recombinant Ad, the number of mRNAs modulated upon infection was significantly reduced but the expression signatures were similar. This reagent provides a valuable tool for studies of Ad in that researchers can examine the effect of infection in the presence of the virion capsid alone.


Assuntos
Adenoviridae/fisiologia , Capsídeo/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , DNA Viral/ultraestrutura , Eletroforese em Gel de Poliacrilamida , Microscopia de Força Atômica , Microscopia Eletrônica , Proteínas Virais/genética , Proteínas Virais/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA