Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(22)2020 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-33213062

RESUMO

Pancreatic cancer remains one of the most difficult malignancies to treat. Minimal improvements in patient outcomes and persistently abysmal patient survival rates underscore the great need for new treatment strategies. Currently, there is intense interest in therapeutic strategies that target tyrosine protein kinases. Here, we employed kinome arrays and bioinformatic pipelines capable of identifying differentially active protein tyrosine kinases in different patient-derived pancreatic ductal adenocarcinoma (PDAC) cell lines and wild-type pancreatic tissue to investigate the unique kinomic networks of PDAC samples and posit novel target kinases for pancreatic cancer therapy. Consistent with previously described reports, the resultant peptide-based kinome array profiles identified increased protein tyrosine kinase activity in pancreatic cancer for the following kinases: epidermal growth factor receptor (EGFR), fms related receptor tyrosine kinase 4/vascular endothelial growth factor receptor 3 (FLT4/VEGFR-3), insulin receptor (INSR), ephrin receptor A2 (EPHA2), platelet derived growth factor receptor alpha (PDGFRA), SRC proto-oncogene kinase (SRC), and tyrosine kinase non receptor 2 (TNK2). Furthermore, this study identified increased activity for protein tyrosine kinases with limited prior evidence of differential activity in pancreatic cancer. These protein tyrosine kinases include B lymphoid kinase (BLK), Fyn-related kinase (FRK), Lck/Yes-related novel kinase (LYN), FYN proto-oncogene kinase (FYN), lymphocyte cell-specific kinase (LCK), tec protein kinase (TEC), hemopoietic cell kinase (HCK), ABL proto-oncogene 2 kinase (ABL2), discoidin domain receptor 1 kinase (DDR1), and ephrin receptor A8 kinase (EPHA8). Together, these results support the utility of peptide array kinomic analyses in the generation of potential candidate kinases for future pancreatic cancer therapeutic development.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neoplasias Pancreáticas/enzimologia , Proteínas Tirosina Quinases/biossíntese , Carcinoma Ductal Pancreático/genética , Humanos , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Proteínas Tirosina Quinases/genética , Proto-Oncogene Mas
2.
Int J Mol Sci ; 21(22)2020 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-33233470

RESUMO

Kinase drug discovery represents an active area of therapeutic research, with previous pharmaceutical success improving patient outcomes across a wide variety of human diseases. In pancreatic ductal adenocarcinoma (PDAC), innovative pharmaceutical strategies such as kinase targeting have been unable to appreciably increase patient survival. This may be due, in part, to unchecked desmoplastic reactions to pancreatic tumors. Desmoplastic stroma enhances tumor development and progression while simultaneously restricting drug delivery to the tumor cells it protects. Emerging evidence indicates that many of the pathologic fibrotic processes directly or indirectly supporting desmoplasia may be driven by targetable protein tyrosine kinases such as Fyn-related kinase (FRK); B lymphoid kinase (BLK); hemopoietic cell kinase (HCK); ABL proto-oncogene 2 kinase (ABL2); discoidin domain receptor 1 kinase (DDR1); Lck/Yes-related novel kinase (LYN); ephrin receptor A8 kinase (EPHA8); FYN proto-oncogene kinase (FYN); lymphocyte cell-specific kinase (LCK); tec protein kinase (TEC). Herein, we review literature related to these kinases and posit signaling networks, mechanisms, and biochemical relationships by which this group may contribute to PDAC tumor growth and desmoplasia.


Assuntos
Adenocarcinoma/genética , Tumor Desmoplásico de Pequenas Células Redondas/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Adenocarcinoma/patologia , Tumor Desmoplásico de Pequenas Células Redondas/patologia , Receptor com Domínio Discoidina 1/genética , Progressão da Doença , Humanos , Neoplasias Pancreáticas/patologia , Proteínas Tirosina Quinases/genética , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-hck/genética , Transdução de Sinais , Quinases da Família src/genética
3.
Surgeon ; 15(1): 24-29, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27374183

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a deadly cancer with an overall 5-year survival rate less than 5% due to the poor early diagnosis and lack of effective therapeutic options. The most effective therapy remains surgery, however post-operative survival could be enhanced with effective adjuvant therapy. The massive information gained from Omics techniques on PDAC at the beginning of the 21st century is a remarkable accomplishment. However, the information gained from the omics data, including next generation sequencing data, has yet to successfully affect care of patients suffering with PDAC. Therefore, we propose the development of an actionable genomic platform that matches a patient's PDAC clinically actionable genes with potential targeted adjuvant therapies. Using this platform, PDX1 has been identified as a potential actionable gene for PDAC, therefore, RNAi therapy, gene therapy and small inhibitory drugs, all targeting PDX1, serve as potential targeted adjuvant therapies. Preclinical studies support the hypothesis that identification of PDAC actionable genes could permit translation of a patient's genomic information into precision targeted adjuvant therapy for PDAC.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Medicina de Precisão , Humanos
4.
J Control Release ; 368: 413-429, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38431093

RESUMO

Exosomes continue to attract interest as a promising nanocarrier drug delivery technology. They are naturally derived nanoscale extracellular vesicles with innate properties well suited to shuttle proteins, lipids, and nucleic acids between cells. Nonetheless, their clinical utility is currently limited by several major challenges, such as their inability to target tumor cells and a high proportion of clearance by the mononuclear phagocyte system (MPS) of the liver and spleen. To overcome these limitations, we developed "Smart Exosomes" that co-display RGD and CD47p110-130 through CD9 engineering (ExoSmart). The resultant ExoSmart demonstrates enhanced binding capacity to αvß3 on pancreatic ductal adenocarcinoma (PDAC) cells, resulting in amplified cellular uptake in in vitro and in vivo models and increased chemotherapeutic efficacies. Simultaneously, ExoSmart significantly reduced liver and spleen clearance of exosomes by inhibiting macrophage phagocytosis via CD47p110-130 interaction with signal regulatory proteins (SIRPα) on macrophages. These studies demonstrate that an engineered exosome drug delivery system increases PDAC therapeutic efficacy by enhancing active PDAC targeting and prolonging circulation times, and their findings hold tremendous translational potential for cancer therapy while providing a concrete foundation for future work utilizing novel peptide-engineered exosome strategies.


Assuntos
Carcinoma Ductal Pancreático , Exossomos , Neoplasias Pancreáticas , Humanos , Exossomos/metabolismo , Antígeno CD47 , Linhagem Celular Tumoral , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia
5.
FASEB J ; 25(8): 2592-603, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21525490

RESUMO

Serum response factor (SRF) is an essential regulator of myogenic and neurogenic genes and the ubiquitously expressed immediate-early genes. The purpose of this study is to determine SRF expression pattern in murine pancreas and examine the role of SRF in pancreatic gene expression. Immunohistochemical analysis of wild-type pancreas and LacZ staining of pancreas from SRF LacZ knock-in animals showed that SRF expression is restricted to ß cells. SRF bound to the rat insulin promoter II (RIP II) serum response element, an element conserved in both rat I and murine I and II insulin promoters. SRF activated RIP II, and SRF binding to RIP II and the exon 5-encoded 64-aa subdomain of SRF was required for this activation. Transient or stable knockdown of SRF leads to down-regulation of insulin gene expression, suggesting that SRF is required for insulin gene expression. Further, SRF physically interacted with the pancreas and duodenum homeobox-1 (Pdx-1) and synergistically activated RIP II. Elevated glucose concentration down-regulated SRF binding to RIP II SRE, and this down-regulation was associated with decreased RIP II activity and increased SRF phosphorylation on serine 103. Together, our results demonstrate that SRF is a glucose concentration-sensitive regulator of insulin gene expression.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/genética , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ratos , Fator de Resposta Sérica/deficiência , Transativadores/metabolismo , Transfecção
6.
Colloids Surf B Biointerfaces ; 218: 112733, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35981473

RESUMO

Effectively capturing, releasing, and reanalyzing circulating tumor cells (CTCs) are critical in cancer diagnosis and individualized treatment. Traditional immunomagnetic separation has disadvantages of low sensitivity and specificity, and is time-consuming and costly in CTCs capture. It is also easily disturbed by the microenvironment in releasing and analyzing CTCs. Here, we proposed an aptamer-mediated DNA concatemer functionalized magnetic nanoparticles (MNPs-AMDC) for the reversible capture and release of CTCs. In this study, aptamers were used both for efficiently capturing CTCs without complicated assembly steps and stimulus-response switch for releasing CTCs with little influence on cellular activity. The MNPs-AMDC was demonstrated to effectively capture (83%) and release CTCs with a good viability rate (92%). Moreover, this device was also tested in clinical blood samples, which would provide a universal tool for diagnosing cancer and treating individuals.


Assuntos
Aptâmeros de Nucleotídeos , Nanopartículas de Magnetita , Células Neoplásicas Circulantes , Linhagem Celular Tumoral , Separação Celular , DNA , Humanos , Magnetismo , Células Neoplásicas Circulantes/patologia , Microambiente Tumoral
7.
World J Surg ; 35(8): 1700-14, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21557010

RESUMO

Normal cellular survival is dependent on the cooperative expression of genes' signaling through a broad array of DNA patterns. Cancer, however, has an Achilles' heel. Its altered cellular survival is dependent on a limited subset of signals through mutated DNA, possibly as few as three. Identification and control of these signals through the use of RNA interference (RNAi) technology may provide a unique clinical opportunity for the management of cancer that employs genomic-proteomic profiling to provide a molecular characterization of the cancer, leading to targeted therapy customized to an individual cancer signal. Such an approach has been described as "personalized therapy." The present review identifies unique developing technology that employs RNAi as a method to target, and therefore block, signaling from mutated DNA and describes a clinical pathway toward its development in cancer therapy.


Assuntos
Genótipo , Neoplasias/genética , Neoplasias/terapia , Medicina de Precisão/tendências , Interferência de RNA/fisiologia , Previsões , Predisposição Genética para Doença/genética , Humanos
8.
World J Surg ; 35(8): 1738-45, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21286716

RESUMO

BACKGROUND: Recent research has focused on signaling cascades and their interactions yielding considerable insight into which genetic pathways are targeted and how they tend to be altered in tumors. Therapeutic interventions now can be designed based on the knowledge of pathways vital to tumor growth and survival. These critical targets for intervention, master switches for cancer, are termed so because the tumor attempts to "flip the switch" in a way that promotes its survival, whereas molecular therapy aims to "switch off" signals important for tumor-related processes. METHODS: Literature review. CONCLUSIONS: Defining useful targets for therapy depends on identifying pathways that are crucial for tumor growth, survival, and metastasis. Because not all signaling cascades are created equal, selecting master switches or targets for intervention needs to be done in a systematic fashion. This discussion proposes a set of criteria to define what it means to be a cancer master switch and provides examples to illustrate their application.


Assuntos
Transformação Celular Neoplásica/genética , Genes de Troca/genética , Neoplasias/genética , Medicina de Precisão/tendências , Transdução de Sinais/genética , Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Neoplasias Colorretais/genética , Análise Mutacional de DNA , Receptores ErbB/genética , Previsões , Proteínas de Homeodomínio/genética , Humanos , Neoplasias Pulmonares/genética , Camundongos , Neoplasias Pancreáticas/genética , Transativadores/genética , Via de Sinalização Wnt/genética
9.
World J Surg ; 35(8): 1715-24, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21249361

RESUMO

BACKGROUND: Somatostatin receptor subtype 5 (SSTR5) mediates the inhibitory effect of somatostatin on insulin expression/secretion and cell proliferation. A number of single nucleotide polymorphisms (SNPs) of SSTR5 have been identified, including P335L, a nonsynonymous SNP located in the protein C-terminal region and encrypted by the codon CCG (proline) or the codon CTG (leucine). In the present study we sought to determine the distribution of the SSTR5 P335L SNP in a cohort of pancreatic cancer patients and whether the P335L SNP affected cellular function of SSTR5 in human pancreatic cancer. METHODS: The P335L germline genotype of 246 patients with pancreatic cancer (213 Caucasians, 16 Hispanics, and 17 African Americans) and 17 human pancreatic cell lines was determined with the TaqMan SNP Genotyping assay. Human SSTR5 leucine variant (L335) was generated by performing site-directed mutagenesis using SSTR5 proline variant (P335) as a template. Transient transfections were performed in HEK293, Mia PaCa-2, and ß-TC-6 cells using Lipofectamine 2000. The expression of SSTR5 L335 was determined with a mouse monoclonal anti-SSTR5 L335 antibody generated in our laboratory. The cell proliferation rate was measured by performing MTS assays. Insulin concentration was measured by performing ELISA assays. RESULTS: Genotyping of the patients' blood indicated that the frequency of the T allele (CT and TT genotypes) in codon 335 of SSTR5 in Caucasians, Hispanics, and African Americans was 52, 69, and 35%, respectively, which was race-dependent. Statistical analysis indicated that association between the frequency of the T allele and the existence of pancreatic cancer in each race missed significance perhaps due to limited sample size. In 17 tested human pancreatic cancer cell lines, 5 (Capan-2, HPAF-II, Panc03.27, Panc-1, and -3) were homozygous (TT genotype) and 9, including Mia PaCa-2, were heterozygous (CT genotype). Overexpression of SSTR5 L335 in Mia PaCa-2 cells enhanced cell proliferation compared to overexpression of SSTR5 P335. Overexpression of SSTR5 P335 enhanced the inhibitory effect of SSTR5 agonist RPL-1980 on cell proliferation of Mia PaCa-2 cells and glucose-stimulated insulin secretion from mouse insulinoma cells, while overexpression of SSTR5 L335 blocked the inhibitory effect of RPL-1980. Overexpression of SSTR5 L335 enhanced PDX-1 expression in Mia PaCa-2 cells. A specific monoclonal antibody was generated to detect SSTR5 P335L. CONCLUSION: SSTR5 P335L SNP widely exists in the human population, in patients with pancreatic cancer, and is race-dependent. The SNP is also present in selected human pancreatic cancer cell lines. In contrast to SSTR5 P335, overexpression of the SSTR5 L335 variant resulted in cellular proliferation and PDX-1 overexpression in human pancreatic cancer cells. Its overexpression blocked the inhibitory effect of an SSTR5-specific analog on human pancreatic cancer cell proliferation and on glucose-stimulated insulin secretion from mouse insulinoma cells. These data suggest that SSTR5 P335L is a hypofunctional protein with a potentially harmful effect on function, as well as potential latent effect, and therefore it could affect the clinical response to somatostatin analog therapy for patients with pancreatic cancer.


Assuntos
Adenocarcinoma/genética , Alelos , Genótipo , Neoplasias Pancreáticas/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores de Somatostatina/genética , Adenocarcinoma/etnologia , Negro ou Afro-Americano/genética , Animais , Anticorpos Monoclonais/genética , Linhagem Celular Tumoral , Proliferação de Células , Códon/genética , Regulação Neoplásica da Expressão Gênica/genética , Frequência do Gene , Hispânico ou Latino/genética , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Pancreáticas/etnologia , Receptores de Somatostatina/imunologia , População Branca/genética
10.
Front Oncol ; 11: 743490, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34707991

RESUMO

OBJECTIVE: To establish a radiomics signature and a nomogram model based on enhanced CT images to predict the Ki-67 index of lung cancer. METHODS: From January 2014 to December 2018, 282 patients with lung cancer who had undergone enhanced CT scans and Ki-67 examination within 2 weeks were retrospectively enrolled and analyzed. The clinical data of the patients were collected, such as age, sex, smoking history, maximum tumor diameter and serum tumor markers. Our primary cohort was randomly divided into a training group (n=197) and a validation group (n=85) at a 7:3 ratio. A Ki-67 index ≤ 40% indicated low expression, and a Ki-67 index > 40% indicated high expression. In total, 396 radiomics features were extracted using AK software. Feature reduction and selection were performed using the lasso regression model. Logistic regression analysis was used to establish a multivariate predictive model to identify high and low Ki-67 expression in lung cancer. A nomogram integrating the radiomics score was established based on multiple logistic regression analysis. Area under the curve (AUC) was used to evaluate the prediction efficiency of the radiomics signature and nomogram. RESULTS: The AUC,sensitivity, specificity and accuracy of the radiomics signature in the training and validation groups were 0.88 (95% CI: 0.82~0.93),79.2%,84.3%,81.2% and 0.86 (95% CI: 0.78~0.94),74.6%,88.1%,79.8%, respectively. A nomogram combining radiomics features and clinical risk factors (smoking history and NSE) was developed. The AUC, sensitivity, specificity and accuracy were 0.87 (95% CI: 0.80~0.95), 75.0%, 90.2% and 83.5% in the validation group, respectively. CONCLUSION: The radiomics signature and nomogram based on enhanced CT images provide a way to predict the Ki-67 expression level in lung cancer.

11.
Cancer Lett ; 491: 97-107, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-32829010

RESUMO

Patients with pancreatic adenocarcinoma (PDAC) have a 5-year survival rate of 8%, the lowest of any cancer in the United States. Traditional chemotherapeutic regimens, such as gemcitabine- and fluorouracil-based regimens, often only prolong survival by months. Effective precision targeted therapy is therefore urgently needed to substantially improve survival. In an effort to expedite approval and delivery of targeted therapy to patients, we utilized a platform to develop a novel combination of FDA approved drugs that would target pancreaticoduodenal homeobox1 (PDX1) and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) utilizing super-promoters of the target genes to interrogate an FDA approved drug library. We identified and selected metformin, simvastatin and digoxin (C3) as a novel combination of FDA approved drugs, which were shown to effectively target PDX1 and BIRC5 in human PDAC tumors in mice with no toxicity.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Digoxina/administração & dosagem , Reposicionamento de Medicamentos , Proteínas de Homeodomínio/antagonistas & inibidores , Metformina/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Sinvastatina/administração & dosagem , Survivina/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Combinação de Medicamentos , Sinergismo Farmacológico , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Camundongos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/patologia
12.
Oncol Rep ; 44(6): 2569-2580, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33125137

RESUMO

Statins, a class of commonly prescribed cholesterol­lowering medications, have been revealed to influence the risk of multiple types of cancer. However, the antitumor effects of statins on pancreatic cancer and their differential efficacy among a variety of statins are not currently well­defined. The aim of the present study was therefore to identify and compare the genes and related biological pathways that were affected by each individual statin on pancreatic cancer. Two human pancreatic cancer cell lines, MiaPaCa2 and PANC1, were exposed to three statins, lovastatin, fluvastatin and simvastatin. The inhibitory effect of statins on pancreatic cancer cell proliferation was first validated. Next, RNA­seq analysis was used to determine the gene expression alterations in either low (2 µM) or high (20 µM) statin concentration­treated cancer cells. Marked differences in gene transcription profiles of both pancreatic cancer cell lines exposed to high concentration statins were observed. Notably, the high concentration statins significantly suppressed core­gene CCNA2­associated cell cycle and DNA replication pathways and upregulated genes involved in ribosome and autophagy pathways. However, the low concentration statin­induced gene expression alterations were only detected in MiaPaCa2 cells. In conclusion, a marked difference in the intra and inter cell­type performance of pancreatic cancer cells exposed to a variety of statins at low or high concentrations was reported herein, which may provide insights for the potential clinical use of statins in future pancreatic cancer therapeutics.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Transcriptoma/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina A2/metabolismo , Replicação do DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Fluvastatina/farmacologia , Fluvastatina/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , RNA-Seq , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico
13.
Exp Ther Med ; 19(1): 347-352, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31853310

RESUMO

In the present study, the risk factors for systemic air embolism as a complication of percutaneous CT-guided lung biopsy were explored. Data from 2,026 percutaneous CT-guided lung biopsy procedures were retrospectively analyzed. All cases were divided into a concurrent air embolism group and a control group, depending on whether air embolism occurred during the puncture process. A systemic air embolism was confirmed when CT values <-200 Hounsfield units were observed in two sequential images. A total of 19 cases (0.9%) of air embolism were detected among the 2,026 patients subjected to percutaneous CT-guided lung biopsy procedures. The most frequently detected embolism site was the left ventricle (89.5%). Only 3 cases (15.8%) were accompanied by obvious clinical symptoms. The results indicated that a puncture location above the level of the left atrium and coughing during the procedure significantly altered the likelihood of embolism developing (P=0.002 and P=0.014 vs. control, respectively). In conclusion, a puncture lesion above the level of the left atrium and coughing during the procedure may be risk factors for air embolism development.

14.
Cancer Lett ; 457: 10-19, 2019 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31059751

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer mortality with a dismal overall survival rate and an urgent need for detection of minute tumors. Current diagnostic modalities have high sensitivity and specificity for larger tumors, but not for minute PDAC. In this study, we test the feasibility of a precision diagnostic platform for detecting and localizing minute human PDAC in mice. This platform includes: 1) defining BIRC5 as an early PDAC-upregulated gene and utilizing an enhanced BIRC5 super-promoter to drive expression of dual Gaussia luciferase (GLuc) and sr39 thymidine kinase (sr39TK) reporter genes exponentially and specifically in PDAC; 2) utilizing a genetically-engineered AAV2RGD to ensure targeted delivery of GLuc and sr39TK specifically to PDAC; 3) using serologic GLuc and sr39TK microPET/CT imaging to detect and localize minute human PDAC in mice. The study demonstrates feasibility of a precision diagnostic platform using an integrated technology through a multiple-stage amplification strategy of dual reporter genes to enhance the specificity and sensitivity of detection and localization of minute PDAC tumors and currently undetectable disease.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/diagnóstico por imagem , Imagem Molecular , Imagem Óptica , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Survivina/metabolismo , Microtomografia por Raio-X , Animais , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Estudos de Viabilidade , Regulação Neoplásica da Expressão Gênica , Humanos , Luciferases/genética , Luciferases/metabolismo , Masculino , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Valor Preditivo dos Testes , Regiões Promotoras Genéticas , Survivina/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Carga Tumoral , Regulação para Cima
15.
World J Gastroenterol ; 14(38): 5823-6, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18855980

RESUMO

AIM: To examine the expression of pancreatic duodenal homeobox-1 (PDX-1) transcription factor in human colorectal cancer. METHODS: RT-PCR, Western blotting, and immuno-histochemistry were performed to determine the expression pattern of transcription factor PDX-1 in primary colorectal tumor, hepatic metastasis, and benign colon tissue from a single patient. RESULTS: The highest PDX-1 transcription levels were detected in the metastasis material. Lower levels of PDX-1 were found to be present in the primary tumor, while normal colon tissue failed to express detectable levels of PDX-1. Western blot data revealed a PDX-1 expression pattern identical to that of mRNA expression. Immunohistochemistry confirmed high metastasis PDX-1 expression, lower levels in the primary tumor, and the presence of only traces of PDX-1 in normal colon tissue. CONCLUSION: These data argue for further evaluation of PDX-1 as a biomarker for colorectal cancer.


Assuntos
Adenocarcinoma/química , Biomarcadores Tumorais/análise , Neoplasias Colorretais/química , Proteínas de Homeodomínio/análise , Neoplasias Hepáticas/química , Transativadores/análise , Adenocarcinoma/genética , Adenocarcinoma/secundário , Adenocarcinoma/terapia , Biomarcadores Tumorais/genética , Quimioterapia Adjuvante , Colectomia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/análise , Transativadores/genética
16.
Cancer Lett ; 418: 75-83, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29309817

RESUMO

Our previous studies have shown that a rat insulin promoter II fragment (RIP) was used to effectively target pancreatic adenocarcinoma (PDAC) and insulinoma that over-express pancreatic and duodenal homeobox-1 (PDX-1). To enhance the activity and specificity of the human insulin promoter, we engineered a synthetic human insulin super-promoter (SHIP). Reporter assay demonstrated that SHIP1 was the most powerful promoter among all of the SHIPs and had far greater activity than the endogenous human insulin promoters and RIP in PDAC expressing PDX-1. Over-expression, knockdown and competitive inhibition of PDX-1 expression assay proved that PDX-1 is a critical transcript factor to regulate the activity of SHIP1. SHIP1-driven viral thymidine kinase followed by ganciclovir (SHIP1-TK/GCV) resulted in cytotoxicity to PDAC cells in vitro. Systemic delivery of SHIP1-TK/GCV in PDAC xenograft mice significantly suppressed PANC-1 tumor growth in vivo greater than RIP-TK/GCV and CMV-TK/GCV controls (p < .05). These preclinical data suggest that SHIP1 is a powerful novel promoter that can be used to target human PDAC expressing PDX-1 in clinical trials. Furthermore, this novel strategy of engineering synthetic super-promoters could be used for other cancer targets.


Assuntos
Adenocarcinoma/genética , Proteínas de Homeodomínio/genética , Insulinas/genética , Neoplasias Pancreáticas/genética , Regiões Promotoras Genéticas/genética , Transativadores/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Animais , Linhagem Celular Tumoral , Ganciclovir/farmacologia , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Células HEK293 , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos SCID , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transativadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
17.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 24(6): 696-8, 2007 Dec.
Artigo em Zh | MEDLINE | ID: mdl-18067087

RESUMO

OBJECTIVE: To investigate the clinical and laboratory characteristics of various hematopoietic malignant patients with t(3;3)(q21;q26) or inv(3) (q21q26). METHODS: Bone marrow samples were collected at presentation, prepared by short-time unstimulated culture and R-binding, and karyotyped by conventional cytogenetical assay (CCA); megalokaryocytes were detected by Streptavidin-AKP (SAP); immunotype of the leukemia cells was tested by flow cytometric anylysis of surface antigens (FACS). RESULTS: All of the 9 hematopoietic malignant patients with t(3;3)(q21;q26) or inv(3) (q21q26) manifested myelodysplasia and poor treatment response. One of them relapsed shortly after allogenic hemotopoietic stem cell transplantation (allo-HSCT). CONCLUSION: Patients with 3q21q26 rearrangement can be found in various hematopoietic malignances and demonstrate an unique entity. These patients show poor treatment response and have extremely poor prognosis.


Assuntos
Cromossomos Humanos Par 3/genética , Rearranjo Gênico , Neoplasias Hematológicas/genética , Cariotipagem , Síndromes Mielodisplásicas/genética , Adulto , Inversão Cromossômica , Mapeamento Cromossômico , Feminino , Neoplasias Hematológicas/patologia , Humanos , Leucemia/genética , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/patologia , Translocação Genética
18.
Zhonghua Yi Xue Za Zhi ; 87(38): 2693-7, 2007 Oct 16.
Artigo em Zh | MEDLINE | ID: mdl-18167247

RESUMO

OBJECTIVE: To study the clinical and laboratory features of myelodysplastic syndromes (MDS) with chromosomal structural changes. METHODS: Among 584 MDS cases with cytogenetic data, 50 patients with chromosomal structural changes, 34 males and 16 females, aged 50.5 (9 approximately 77), were reclassified according to the WHO criteria, and their clinical and laboratory features were analyzed retrospectively. RESULTS: The incidence of chromosomal structural changes in the MDS patients was 7.4%. i (17) (q10), t (1; 3) (p36; q21), der (1; 7) (q10; p10), and der (22) occurred frequently. The chromosomal structural changes in 13 cases were reported in the literatures for the first time. The patients with i (17) (q10) were characterized by moderate to severe anemia and a poor prognosis. Predominant dysgranulocytopoiesis and dysmegakaryocytopoiesis, including a nuclear shift to the left, pseudo-Pelger-Hüet anomaly, hypogranularity, and increased micromegakaryocytes. The patients with t (1; 3) (p36; q21) revealed macrocytic anemia, obvious dysmegakaryocytopoiesis, and dysgranulocytopoiesis, accompanied by defective differentiation and monocytosis. The bone marrow cells from the MDS patients with t (1; 3) (p36; q21) mainly or only expressed MEL1. The initial symptom of the patients with der (1; 7) (q10; p10) was infection. These patients showed macrocytic or normocytic anemia. Trilineage dysplasia was found in the bone marrow smears. The patients had short median survival. The patients with der (22) revealed anemia, and normal or elevated platelet counts. Hypogranularity and pseudo-Pelger-Hüet anomaly were present in all cases with der (22). The megakaryocytes were small and generally contained one or two nuclei. A translocation involvement of 22q11 was frequently found in the patients with der (22). CONCLUSION: MDS patients with i (17) (q10), t (1; 3) (p36; q21), and der (1; 7) (q10; p10) may be a new unique clinical-pathologic subsets. Whether the MDS patients with der (22) can be considered as a new unique subject remains to be confirmed by future studies.


Assuntos
Aberrações Cromossômicas , Síndromes Mielodisplásicas/genética , Adolescente , Adulto , Idoso , Criança , Estruturas Cromossômicas/genética , Feminino , Seguimentos , Humanos , Cariotipagem , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/diagnóstico , Prognóstico , Estudos Retrospectivos
19.
Chin Med Sci J ; 21(2): 99-103, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16845796

RESUMO

OBJECTIVE: To investigate the role of the burden of abnormal hematopoietic clone in the development of myelodysplastic syndromes (MDS). METHODS: The ratio of the bone marrow cells with abnormal chromosomes to the total counted bone marrow cells was regarded as the index of MDS clone burden. The disease severity related parameters including white blood cell count, hemoglobin, platelet count, lactate dehydrogenase level, bone marrow blast, myeloid differentiation index, micromegakaryocyte, transfusion, interleukin-2, tumor necrosis factor (TNF), CD4+ and CD8+ T cells of MDS patients were assayed, and the correlations between those parameters and MDS clone burden were also analyzed. RESULTS: The clone burden of MDS patients was 67.4% +/- 36.2%. MDS clone burden positively correlated with bone marrow blasts (r = 0.483, P < 0.05), negatively with hemoglobin level (r = -0.445, P < 0.05). The number of blasts, hemoglobin, and erythrocytes in high clone burden (> 50%) and low clone burden ( < or = 50%) groups were 7.78% +/- 5.51% and 3.45% +/- 3.34%, 56.06 +/- 14.28 g/L and 76.40 +/- 24.44 g/L, (1.82 +/- 0.48) x 10(12)/L and (2.32 +/- 0.66) x 10(12)/L, respectively (all P < 0.05). CD4+ T lymphocytes of MDS patients and normal controls were (0.274 +/- 0.719) x 10(9)/L and (0.455 +/- 0.206) x 10(9)/L, respectively (P < 0.05). CD8+ T lymphocytes of MDS patients and normal controls were (0.240 +/- 0.150) x 10(9)/L and (0.305 +/- 0.145) x 10(9)/L, respectively. The serum level of interleukin-2 of MDS patients (6.29 +/- 3.58 ng/mL) was significantly higher than normal control (3.11 +/- 1.40 ng/mL, P < 0.05). The serum level of TNF of MDS patients and normal control group were 2.42 +/- 1.79 ng/mL and 1.68 +/- 0.69 ng/mL, respectively. The ratio of CD4 to CD8 was higher in high clone burden MDS patients (1.90 +/- 0.52) than that in low clone burden patients (0.97 +/- 0.44, P < 0.05). CONCLUSION: The quantitive clonal karyotype abnormalities and deficient T cell immunity are important parameters for evaluating MDS severity and predicting its progression.


Assuntos
Células da Medula Óssea/patologia , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Aberrações Cromossômicas , Feminino , Hematopoese/genética , Células-Tronco Hematopoéticas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/sangue , Células-Tronco Neoplásicas/patologia , Policitemia/genética , Policitemia/patologia , Subpopulações de Linfócitos T/patologia , Adulto Jovem
20.
Zhonghua Nei Ke Za Zhi ; 45(11): 918-21, 2006 Nov.
Artigo em Zh | MEDLINE | ID: mdl-17313880

RESUMO

OBJECTIVE: To investigate the cytogenetic and prognostic significance of acute myeloid leukemia (AML) with t (8; 21). METHODS: 189 patients with t (8; 21) AML were categorized according to their additional karyotypic aberration and their clinical outcomes analysed. RESULTS: Among them, 63 patients (33.3%) were t (8; 21) without other additional aberrations, 126 cases (66.7%) were t (8; 21) with other additional aberrations. -Y was found in 46.7% (63/135) of the male and -X was found in 25.9% (14/54) of female patients. In additional aberrations, loss of the sex chromosome were found in 77 cases (61.1%), Del (9q) was found in 16 cases (12.7%), +4 was found in 5 cases (4.0%); 7q- was found in 6 cases (4.8%); Tetraploidy (4N) was found in 2 cases (1.6%); Variant translocation was found in 7 cases (5.6%). The 189 patients had a high remission rate (87.0%) and a relatively long median survival (21.6 months). +4 and 4N were an unfavorable prognostic factors. Fluorescence in situ hybridization technique is a more sensitive and accurate method to detect t (8; 21), especially in variant translocation, complex variant translocation and masked translocation. CONCLUSION: t (8; 21) AML is also frequently associated with additional chromosome aberrations, these aberration had influence on prognosis.


Assuntos
Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 8/genética , Leucemia Mieloide Aguda/genética , Adolescente , Adulto , Idoso , Citogenética , Feminino , Seguimentos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Masculino , Pessoa de Meia-Idade , Prognóstico , Translocação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA