Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Tissue Bank ; 22(4): 727-736, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34432159

ABSTRACT

Biobanks play a crucial role in enabling biomedical research by facilitating scientific use of valuable human biomaterials. The PALGA foundation-a nationwide network and registry of histo- and cytopathology in the Netherlands-was established to promote the provision of data within and between pathology departments, and to make the resulting knowledge available for healthcare. Apart from the pathology data, we aimed to utilize PALGA's nationwide network to find and access the rich wealth of Formalin-Fixed Paraffin-Embedded (FFPE) tissue samples for scientific use.  We implemented the Dutch National TissueArchive Portal (DNTP) to utilize PALGA's nationwide network for requesting FFPE tissue samples. The DNTP consists of (1) a centrally organized internet portal to improve the assessing, processing, harmonization, and monitoring of the procurement process, while (2) dedicated HUB-employees provide practical support at peripheral pathology departments. Since incorporation of the DNTP, both the number of filed requests for FFPE tissue samples and the amount of HUB-mediated support increased 55 and 29% respectively. In line, the sample procurement duration time decreased significantly (- 47%). These findings indicate that implementation of the DNTP improved the frequency, efficiency, and transparency of FFPE tissue sample procurement for research in the Netherlands. To conclude, the need for biological resources is growing persistently to enable precision medicine. Here, we access PALGA's national, pathology network by implementation of the DNTP to allow for efficient, consistent, and transparent exchange of FFPE tissue samples for research across the Netherlands.


Subject(s)
Biomedical Research , Humans , Netherlands
2.
Biochim Biophys Acta ; 1834(11): 2242-58, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23376433

ABSTRACT

Despite major improvements on the knowledge and clinical management, cancer is still a deadly disease. Novel biomarkers for better cancer detection, diagnosis and treatment prediction are urgently needed. Proteins secreted, shed or leaking from the cancer cell, collectively termed the cancer secretome, are promising biomarkers since they might be detectable in blood or other biofluids. Furthermore, the cancer secretome in part represents the tumor microenvironment that plays a key role in tumor promoting processes such as angiogenesis and invasion. The cancer secretome, sampled as conditioned medium from cell lines, tumor/tissue interstitial fluid or tumor proximal body fluids, can be studied comprehensively by nanoLC-MS/MS-based approaches. Here, we outline the importance of current cancer secretome research and describe the mass spectrometry-based analysis of the secretome. Further, we provide an overview of cancer secretome research with a focus on the three most common cancer types: lung, breast and colorectal cancer. We conclude that the cancer secretome research field is a young, but rapidly evolving research field. Up to now, the focus has mainly been on the discovery of novel promising secreted cancer biomarker proteins. An interesting finding that merits attention is that in cancer unconventional secretion, e.g. via vesicles, seems increased. Refinement of current approaches and methods and progress in clinical validation of the current findings are vital in order to move towards applications in cancer management. This article is part of a Special Issue entitled: An Updated Secretome.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Colon/pathology , Colorectal Neoplasms/pathology , Lung Neoplasms/pathology , Lung/pathology , Proteome/metabolism , Rectum/pathology , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Breast/metabolism , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Colon/metabolism , Colorectal Neoplasms/blood , Colorectal Neoplasms/metabolism , Female , Humans , Lung/metabolism , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Mass Spectrometry/methods , Proteome/analysis , Proteomics/methods , Rectum/metabolism , Secretory Pathway , Tumor Microenvironment
3.
J Proteomics ; 74(4): 558-66, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21262401

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common type of cancer worldwide. Strong prognostic indicators that predict development of distant metastases are the presence and number of lymph node metastases in the neck, and extranodal spread. Recently, it was shown in several studies that also the presence of disseminated tumor cells (DTC) in the bone marrow predicts development of distant metastases. We have investigated whether protein signatures could be detected in primary HNSCC that distinguish tumors that disseminate into the bone marrow from those that do not. Therefore, DTC-positive and -negative primary HNSCC tumors were analyzed by 2D-DIGE. A signature consisting of 51 differential protein spots was identified upon stratification for bone marrow status, which allowed a correct classification of DTC-positive and DTC-negative HNSCC tumors in 95% of cases, using hierarchical clustering. The most prominent feature within this signature was the down-regulation of CK19 in DTC-positive tumors. Our data show that tumor cell dissemination to the bone marrow, the onset of hematogenic metastasis, can be deduced from the protein profile in the primary tumor. The highly significant down-regulation of CK19 supports a model of epithelial-mesenchymal transition for tumors that show a high proclivity for hematogenic dissemination.


Subject(s)
Metabolome , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma/diagnosis , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Squamous Cell , Case-Control Studies , Down-Regulation , Epithelial-Mesenchymal Transition/physiology , Female , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Keratin-19/metabolism , Male , Metabolome/physiology , Middle Aged , Neoplasm Invasiveness , Neoplasms, Squamous Cell/diagnosis , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , Neoplastic Cells, Circulating/pathology , Prognosis , Squamous Cell Carcinoma of Head and Neck , Two-Dimensional Difference Gel Electrophoresis
4.
Int J Cancer ; 128(7): 1596-605, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-20499310

ABSTRACT

A subgroup of head and neck squamous cell carcinomas (HNSCCs) contains high-risk human papillomavirus-type 16 (HPV16). The viral E6 and E7 oncoproteins inactivate the p53 and pRb proteins, respectively. We examined the causative effect of HPV16 E6 and E7 expression on the immortalization of normal oral keratinocytes (OKCs) and compared the resulting phenotype with alternative ways of p53- and pRb-pathway abrogation frequently found in HNSCCs without HPV. Primary OKCs were conditionally immortalized with temperature-sensitive SV40 large T-antigen and human telomerase, allowing these cells to return to their senescent primary state after temperature shift. HPV16 E6 and E7 were introduced to overcome senescence, determined with population doubling (PD) as read-out. For comparison, we downregulated p53 and p16 by short hairpin RNA genes and expressed mutant p53R(175)H and cyclinD1. Expression of HPV16 E6 caused an extended life span similar to expression of mutant p53R(175)H or p53 knockdown. Expression of mutant p53R(175)H seemed to cause additional activation of the hypoxia and WNT signaling pathways. HPV16 E7 expression had no direct effect on lifespan, similar to p16 knockdown or cyclinD1 expression. In combination with HPV16 E6 or other functional inactivations of p53, abrogation of the pRb-pathway by either HPV16 E7 or other manipulations caused an immortal phenotype. Our data show the causative role of HPV16 E6/E7 in early squamous carcinogenesis. Activity of each gene could be mimicked by other genetic events frequently found in HNSCC without HPV. This data provides the experimental proof of causal association of HPV in HNSCC carcinogenesis and further support the crucial role of the p53- and pRb-pathways.


Subject(s)
Gene Expression Regulation , Head and Neck Neoplasms/metabolism , Keratinocytes/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Calcium/metabolism , Cell Line, Tumor , Cyclin D1/biosynthesis , Gene Expression Profiling , Genetic Vectors , Humans , Keratinocytes/cytology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Phenotype , Repressor Proteins/metabolism , Signal Transduction
5.
J Proteomics ; 73(10): 1790-803, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20139037

ABSTRACT

Squamous cell carcinoma of the head and neck (HNSCC) is the sixth most common cancer worldwide. Despite improvements in diagnosis and treatment, the five-year-survival rate of advanced HNSCC has only moderately increased, which is largely due to the high proportion of patients that present with advanced disease stage and the frequent development of relapse and second primary tumors. Protein biomarkers allowing early detection of primary HNSCC or relapse may aid to improve clinical outcome. Screening for precursor changes in the mucosal linings preceding the development of invasive tumors and for accurate prediction of risk of malignant transformation, may be propitious opportunities, which are as yet difficult. This review summarizes recent results in HNSCC proteomics for biomarker research. Despite the wide diversity of experimental designs, a few common markers have been detected. Although some of these potential biomarkers are very promising, they still have to be further clinically validated. Finally, treatment of advanced cancers of several sites within the head and neck has shifted significantly during the last decade, and also, targeted drugs have entered the clinic. This has major consequences for the research questions in HNSCC research and accordingly for the future direction of proteome research in HNSCC biomarker discovery.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/diagnosis , Head and Neck Neoplasms/diagnosis , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Humans , Lymphatic Metastasis/diagnosis , Proteome/genetics , Proteomics/methods
6.
J Proteome Res ; 9(1): 485-94, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19877719

ABSTRACT

Meningiomas are among the most frequent tumors of the brain and spinal cord accounting for 15-20% of all central nervous system tumors and frequently associated with neurofibromatosis type 2. In this study, we aimed to unravel molecular meningioma tumorigenesis and discover novel protein biomarkers for diagnostic and/or prognostic purposes and performed in-depth proteomic profiling of meningioma cells compared to human primary arachnoidal cells. We isolated proteins from meningioma cell line SF4433 and human primary arachnoidal cells and analyzed the protein profiles by Gel-nanoLC-MS/MS in conjunction with protein identification and quantification by shotgun nanoLC tandem mass spectrometry and spectral counting. Differential analysis of meningiomas revealed changes in the expression levels of 281 proteins (P < 0.01) associated with various biological functions such as DNA replication, recombination, cell cycle, and apoptosis. Among several interesting proteins, we focused on a subset of the highly significantly up-regulated proteins, the minichromosome maintenance (MCM) family. We performed subsequent validation studies by qRT-PCR in human meningioma tissue samples (WHO grade I, 14 samples; WHO grade II, 7 samples; and WHO grade III, 7 samples) compared to arachnoidal tissue controls (from fresh autopsies; 3 samples) and found that MCMs are highly and significantly up-regulated in human meningioma tumor samples compared to arachnoidal tissue controls. We found a significant increase in MCM2 (8 fold), MCM3 (5 fold), MCM4 (4 fold), MCM5 (4 fold), MCM6 (3 fold), and MCM7 (5 fold) expressions in meningiomas. This study suggests that MCM family proteins are up-regulated in meningiomas and can be used as diagnostic markers.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Meningioma/metabolism , Nuclear Proteins/metabolism , Proteomics/methods , Algorithms , Arachnoid/metabolism , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
7.
Oral Oncol ; 46(2): 123-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20036603

ABSTRACT

Oral leukoplakia is defined as a white patch in the oral cavity that cannot be diagnosed as any other known disorder. These lesions carry an increased risk of malignant progression, and approximately 2-3% per year do progress to cancer. At present biopsies are histopathologically graded for dysplasia to assess the risk of progression, but this grading is somewhat subjective and of limited use for the individual patient. In a previous study we discovered by a comprehensive proteomics approach that compared to normal mucosa, protein expression of cornulin, keratin 4 and keratin 13 is decreased in tumors and severe dysplasia, preneoplastic tissue with a high risk of malignant progression. Here, we studied whether loss of expression of these proteins can predict malignant transformation of oral leukoplakia. Biopsies of 12 progressing and 36 non-progressing leukoplakia lesions were analyzed for cornulin, keratin 4 and keratin 13 expression by immunohistochemistry, and graded for dysplasia. Kaplan-Meier analysis showed that loss of expression of neither cornulin (p=0.075), keratin 4 (p=0.789) nor keratin 13 (p=0.732) was significantly associated with malignant transformation of leukoplakia lesions. However, decreased expression of these proteins was significantly associated with the presence of hyperkeratosis. Only dysplasia grading correlated significantly with malignant progression of leukoplakia (p=0.024). Despite the promising outlook that decreased cornulin, keratin 4 and keratin 13 expression in the oral mucosa is associated with a premalignant state, these markers do not predict malignant transformation of leukoplakia lesions. The most likely explanation is that the aberrant differentiation state of hyperkeratotic leukoplakia lesions already causes a decreased expression, obscuring the putative association with malignant transformation. Our results support the significance of dysplasia grading for the prediction of malignant transformation.


Subject(s)
Carcinoma, Squamous Cell , Epithelial Cells , Leukoplakia, Oral , Mouth Mucosa , Mouth Neoplasms , Precancerous Conditions , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Keratin-13/metabolism , Keratin-4/metabolism , Leukoplakia, Oral/metabolism , Leukoplakia, Oral/pathology , Male , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prognosis , Risk Factors , Tumor Suppressor Protein p53/metabolism
8.
Clin Cancer Res ; 15(24): 7666-7675, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19996216

ABSTRACT

PURPOSE: The 5-year survival rates of head and neck squamous cell carcinomas (HNSCC) remain disappointing. HNSCCs develop in precursor fields of genetically altered cells that are often not completely resected when the tumor is excised, causing local relapse. These precursor fields are mostly recognized as dysplasia, but histologic grading cannot reliably predict malignant transformation. Our aim was to discover and validate protein biomarkers that can detect precursor fields and predict local relapse in HNSCC using immunostaining of surgical margins. EXPERIMENTAL DESIGN: We compared paired and genetically characterized normal, precursor, and tumor tissues of eight patients by proteome analysis to identify differentially expressed proteins. The prognostic value of candidate protein biomarkers was evaluated by immunohistochemical analysis of 222 surgical margins of 46 HNSCC patients who developed local relapse or remained disease free. Significant associations were determined by Kaplan-Meier survival analysis and Cox-proportional hazards models. RESULTS: Forty proteins showed significant differential expression (false discovery rate-corrected P < 0.05). Most discriminative markers suited for immunostaining were keratin 4 and cornulin. Low expression in the surgical margins of keratin 4 (hazard ratio, 3.8; P = 0.002), cornulin (hazard ratio, 2.7; P = 0.025), and their combination (hazard ratio, 8.8; P = 0.0005) showed a highly significant association with the development of local relapse. Dysplasia grading had no prognostic relevance. CONCLUSIONS: Immunohistochemical assessment of keratin 4 and cornulin expression in surgical margins of HNSCC patients outperforms histopathologic grading in predicting the risk for local relapse. These markers can be used to initiate more frequent and lifelong surveillance of patients at high risk of local relapse, and enable selection for adjuvant treatment or tertiary prevention trials. (Clin Cancer Res 2009;15(24):7666-75).

9.
J Proteomics ; 72(5): 803-14, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19446051

ABSTRACT

The 5-year-survival rate of head and neck squamous cell carcinoma (HNSCC) has been only moderately improved over the last few decades. HNSCC develops in precursor fields of genetically altered mucosal cells, typically characterized by p53 pathway disruption, that mostly do not give any clinical symptoms. Patients present therefore often with invasive carcinomas in an advanced stage. After tumor resection, part of these fields frequently stays behind unnoticed, causing secondary tumors. Identification of these precursor fields would allow screening and early detection of both primary and secondary tumors. Our aim was to identify differential proteins related to p53 dysfunction. These proteins may serve as valuable biomarkers that can predict the presence of a precursor field. We used a squamous cell model for p53 inactivation, which was analyzed by 2D-DIGE and LC-MS/MS. This approach enabled us to identify a set of 74 proteins that were differentially expressed in cells with normal versus disrupted p53 function. For six proteins the major changes in expression were verified with immunohistochemical staining. The most promising result was the identification of peroxiredoxin-1 which allowed immunohistochemical discrimination between normal epithelium and precursor field tissue with a TP53 mutation.


Subject(s)
Carcinogens/metabolism , Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Biomarkers, Tumor/metabolism , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Humans , Immunohistochemistry/methods , Keratinocytes/metabolism , Models, Biological , Mutation , Proteome , Proteomics/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...