Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Cell Proteomics ; 12(2): 381-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23172894

ABSTRACT

Breast cancer is a very heterogeneous disease, encompassing several intrinsic subtypes with various morphological and molecular features, natural history and response to therapy. Currently, molecular targeted therapies are available for estrogen receptor (ER)(-) and human epidermal growth factor receptor 2 (Her2)-positive breast tumors. However, a significant proportion of primary breast cancers are negative for ER, progesterone receptor (PgR), and Her2, comprising the triple negative breast cancer (TNBC) group. Women with TNBC have a poor prognosis because of the aggressive nature of these tumors and current lack of suitable targeted therapies. As a consequence, the identification of novel relevant protein targets for this group of patients is of great importance. Using a systematic two dimensional (2D) gel-based proteomic profiling strategy, applied to the analysis of fresh TNBC tissue biopsies, in combination with a three-tier orthogonal technology (two dimensional PAGE/silver staining coupled with MS, two dimensional Western blotting, and immunohistochemistry) approach, we aimed to identify targetable protein markers that were present in a significant fraction of samples and that could define therapy-amenable sub-groups of TNBCs. We present here our results, including a large cumulative database of proteins based on the analysis of 78 TNBCs, and the identification and validation of one specific protein, Mage-A4, which was expressed in a significant fraction of TNBC and Her2-positive/ER negative lesions. The high level expression of Mage-A4 in the tumors studied allowed the detection of the protein in the tumor interstitial fluids as well as in sera. The existence of immunotherapeutics approaches specifically targeting this protein, or Mage-A protein family members, and the fact that we were able to detect its presence in serum suggest novel management options for TNBC and human epidermal growth factor receptor 2 positive/estrogen receptor negative patients bearing Mage-A4 positive tumors.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Breast Neoplasms/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/blood , Proteome/metabolism , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Carcinoma/diagnosis , Carcinoma/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Humans , Mass Spectrometry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteome/chemistry , Proteome/genetics , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics
2.
Mol Oncol ; 4(6): 539-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21036680

ABSTRACT

Our limited understanding of the biological impact of the whole spectrum of early breast lesions together with a lack of accurate molecular-based risk criteria for the diagnosis and assignment of prognostic significance to biopsy findings presents an important problem in the clinical management of patients harboring precancerous breast lesions. As a result, there is a need to identify biomarkers that can better determine the outcome of early breast lesions by identifying subpopulations of cells in breast premalignant disease that are at high-risk of progression to invasive disease. A first step towards achieving this goal will be to define the molecular phenotypes of the various cell types and precursors - generated by the stem cell hierarchy - that are present in normal and benign conditions of the breast. To date there have been very few systematic proteomic studies aimed at characterizing the phenotypes of the different cell subpopulations present in normal human mammary tissue, partly due to the formidable heterogeneity of mammary tissue, but also due to limitations of the current proteomic technologies. Work in our laboratories has attempted to address in a systematic fashion some of these limitations and here we present our efforts to search for biomarkers using normal fresh tissue from non-neoplastic breast samples. From the data generated by the 2D gel-based proteomic profiling we were able to compile a protein database of normal human breast epithelial tissue that was used to support the biomarker discovery program. We review and present new data on the putative cell-progenitor marker cytokeratin 15 (CK15), and describe a novel marker, dihydropyriminidase-related protein 3 (DRP3) that in combination with CK15 and other well known proteins were used to define molecular phenotypes of normal human breast epithelial cells and their progenitors in resting acini, lactating alveoli, and large collecting ducts of the nipple. Preliminary results are also presented concerning DRP3 positive usual ductal hyperplasias (UDHs) and on single cell layer columnar cells (CCCs). At least two bona fide biomarkers of undifferentiated ERα/PgR negative luminal cells emerged from these studies, CK15 and c-KIT, which in combination with transformation markers may lead to the establishment of a protein signature able to identify breast precancerous at risk of progressing to invasive disease.


Subject(s)
Biomarkers/metabolism , Breast Neoplasms , Epithelial Cells/metabolism , Mammary Glands, Human , Phenotype , Proteomics/methods , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Databases, Protein , Electrophoresis, Gel, Two-Dimensional/methods , Female , Humans , Immunohistochemistry , Immunophenotyping , Keratin-15/metabolism , Keratin-19/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mass Spectrometry/methods , Muscle Proteins/metabolism , Protein Array Analysis/methods , Protein Isoforms/metabolism
3.
Mol Oncol ; 4(1): 65-89, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20005186

ABSTRACT

Breast cancer is by far the most common diagnosed form of cancer and the leading cause of cancer death in women today. Clinically useful biomarkers for early detection of breast cancer could lead to a significant reduction in mortality. Here we describe a detailed analysis using gel-based proteomics in combination with mass spectrometry and immunohistochemistry (IHC) of the tumour interstitial fluids (TIF) and normal interstitial fluids (NIF) collected from 69 prospective breast cancer patients. The goal of this study was to identify abundant cancer up-regulated proteins that are externalised by cells in the tumour microenvironment of most if not all these lesions. To this end, we applied a phased biomarker discovery research strategy to the analysis of these samples rather than comparing all samples among each other, with inherent inter and intra-sample variability problems. To this end, we chose to use samples derived from a single tumour/benign tissue pair (patient 46, triple negative tumour), for which we had well-matched samples in terms of epithelial cell numbers, to generate the initial dataset. In this first phase we found 110 proteins that were up-regulated by a factor of 2 or more in the TIF, some of which were confirmed by IHC. In the second phase, we carried out a systematic computer assisted analysis of the 2D gels of the remaining 68 TIF samples in order to identify TIF 46 up-regulated proteins that were deregulated in 90% or more of all the available TIFs, thus representing common breast cancer markers. This second phase singled out a set of 26 breast cancer markers, most of which were also identified by a complementary analysis using LC-MS/MS. The expression of calreticulin, cellular retinoic acid-binding protein II, chloride intracellular channel protein 1, EF-1-beta, galectin 1, peroxiredoxin-2, platelet-derived endothelial cell growth factor, protein disulfide isomerase and ubiquitin carboxyl-terminal hydrolase 5 were further validated using a tissue microarray containing 70 malignant breast carcinomas of various grades of atypia. A significant number of these proteins have already been detected in the blood/plasma/secretome by others. The next steps, which include biomarker prioritization based on the hierarchal evaluation of these markers, antibody and antigen development, assay development, analytical validation, and preliminary testing in the blood of healthy and breast cancer patients, are discussed.


Subject(s)
Biomarkers, Tumor/metabolism , Biomarkers/metabolism , Breast Neoplasms/diagnosis , Neoplasm Staging/classification , Body Fluids/chemistry , Breast Neoplasms/metabolism , Calreticulin/metabolism , Chloride Channels/metabolism , Early Diagnosis , Electrophoresis, Gel, Two-Dimensional/methods , Female , Galectin 1/metabolism , Humans , Immunohistochemistry , Neoplasm Staging/standards , Peptide Elongation Factor 1/metabolism , Peroxiredoxins/metabolism , Prognosis , Protein Disulfide-Isomerases/metabolism , Proteomics/statistics & numerical data , Thymidine Phosphorylase/metabolism , Up-Regulation
4.
Mol Oncol ; 3(3): 220-37, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19393583

ABSTRACT

Invasive apocrine carcinomas (IACs), as defined by morphological features, correspond to 0.3-4% of all invasive ductal carcinomas (IDC), and despite the fact that they are histologically distinct from other breast lesions there are currently no standard molecular criteria available for their diagnosis and no unequivocal information as to their prognosis. In an effort to address these concerns we have been using protein expression profiling technologies in combination with mass spectrometry and immunohistochemistry (IHC) to discover specific biomarkers that could allow us to molecularly characterize these lesions as well as to dissect some of the steps in the processes underlying breast apocrine metaplasia and development of precancerous apocrine lesions. Establishing these apocrine-specific markers as best practice for the routine pathology evaluation of breast cancer, however, will require their validation in large cohorts of patients. Towards this goal we have composed a panel of antibodies against components of an apocrine protein signature that includes probes against the apocrine-specific markers 15-prostaglandin dehydrogenase (15-PGDH), and acyl-CoA synthetase medium-chain family member 1 (ACSM1), in addition to a set of categorizing markers that are consistently expressed (AR, CD24) or not expressed (ERα, PgR, Bcl-2, and GATA-3) by apocrine metaplasia in benign breast lesions and apocrine sweat glands. This panel was used to analyze a well-defined cohort consisting of 14 apocrine ductal carcinoma in situ (ADCIS), and 33 IACs diagnosed at the Cancer Institute Hospital, Tokyo between 1997 and 2001. Samples were originally classified on the basis of cellular morphology with all cases having more than 90% of the tumour cells exhibiting cytological features typical of apocrine cells. Using the expression of 15-PGDH and/or ACSM1 as the main criterion, but taking into account the expression of other markers, we were able to identify unambiguously 13 out of 14 ADCIS (92.9%) and 20 out of 33 (60.6%) IAC samples, respectively, as being of apocrine origin. Our results demonstrate that IACs correspond to a distinct, even if heterogeneous, molecular subgroup of breast carcinomas that can be readily identified in an unbiased way using a combination of markers that recapitulate the phenotype of apocrine sweat glands (15-PGDH(+), ACSM1(+), AR(+), CD24(+), ERα(-), PgR(-), Bcl-2(-), and GATA-3(-)). These results pave the way for addressing issues such as prognosis of IACs, patient stratification for targeted therapeutics, as well as research strategies for identifying novel therapeutic targets for developing new cancer therapies.


Subject(s)
Apocrine Glands/metabolism , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Apocrine Glands/pathology , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Middle Aged , Retrospective Studies
5.
Mol Cell Proteomics ; 7(10): 1795-809, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18632593

ABSTRACT

Established histopathological criteria divide invasive breast carcinomas into defined groups. Ductal of no specific type and lobular are the two major subtypes accounting for around 75 and 15% of all cases, respectively. The remaining 10% include rarer types such as tubular, cribriform, mucinous, papillary, medullary, metaplastic, and apocrine breast carcinomas. Molecular profiling technologies, on the other hand, subdivide breast tumors into five subtypes, basal-like, luminal A, luminal B, normal breast tissue-like, and ERBB2-positive, that have different prognostic characteristics. An additional subclass termed "molecular apocrine" has recently been described, but these lesions did not exhibit all the histopathological features of classical invasive apocrine carcinomas (IACs). IACs make up 0.5-3% of the invasive ductal carcinomas, and despite the fact that they are morphologically distinct from other breast lesions, there are presently no standard molecular criteria available for their diagnosis and as a result no precise information as to their prognosis. Toward this goal our laboratories have embarked in a systematic proteomics endeavor aimed at identifying biomarkers that may characterize and subtype these lesions as well as targets that may lead to the development of novel targeted therapies and chemoprevention strategies. By comparing the protein expression profiles of apocrine macrocysts and non-malignant breast epithelial tissue we have previously reported the identification of a few proteins that are specifically expressed by benign apocrine lesions as well as by the few IACs that were available to us at the time. Here we reiterate our strategy to reveal apocrine cell markers and present novel data, based on the analysis of a considerably larger number of samples, establishing that IACs correspond to a distinct molecular subtype of breast carcinomas characterized by the expression of 15-prostaglandin dehydrogenase alone or in combination with a novel form of acyl-CoA synthetase medium-chain family member 1 (ACSM1). Moreover we show that 15-prostaglandin dehydrogenase is not expressed by other breast cancer types as determined by gel-based proteomics and immunohistochemistry analysis and that antibodies against this protein can identify IACs in an unbiased manner in a large breast cancer tissue microarray making them potentially useful as a diagnostic aid.


Subject(s)
Apocrine Glands/enzymology , Apocrine Glands/pathology , Breast Neoplasms/classification , Breast Neoplasms/enzymology , Coenzyme A Ligases/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Cohort Studies , Disease Progression , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Immunophenotyping , Middle Aged , Neoplasm Invasiveness , Paraffin Embedding , Phenotype , Silver Staining , Tissue Array Analysis
6.
Int J Cancer ; 121(7): 1433-44, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17565747

ABSTRACT

The S100A4 protein, which is involved in the metastasis process, is a member of the S100 superfamily of Ca-binding proteins. Members of this family are multifunctional signaling proteins with dual extra and intracellular functions involved in the regulation of diverse cellular processes. Several studies have established a correlation between S100A4 protein expression and worse prognosis for patients with various malignancies including breast cancer. In this article, we have used specific antibodies in combination with immunohistochemistry (IHC) to identify the cell types that express S100A4 in human breast cancer biopsies obtained from high-risk patients. IHC analysis of 68 tumor biopsies showed that the protein is expressed preferentially by various cell types present in the tumor microenvironment (macrophages, fibroblasts, activated lymphocytes), rather than by the tumor cells themselves. Moreover, we show that the protein is externalized by the stroma cells to the fluid that bathes the tumor microenvironment, where it is found in several forms that most likely correspond to charge variants. Using a specific ELISA test, we detected a significant higher concentration of S100A4 in the tumor interstitial fluid (TIF) as compared to their corresponding normal counterparts (NIF).


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , S100 Proteins/biosynthesis , Adult , Aged, 80 and over , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/chemistry , Fibroblasts/pathology , Humans , Immunohistochemistry/methods , Lymphocytes/chemistry , Lymphocytes/pathology , Macrophages/chemistry , Macrophages/pathology , Middle Aged , S100 Calcium-Binding Protein A4 , S100 Proteins/blood , S100 Proteins/immunology
7.
Mol Oncol ; 1(1): 97-119, 2007 Jun.
Article in English | MEDLINE | ID: mdl-19383289

ABSTRACT

The identification as well as the molecular characterization of breast precancerous lesions in terms of increased risk of progression and/or recurrence is becoming a critical issue today as improved non-surgical procedures are detecting cancer at an earlier stage. The strategy we have been pursuing to identify early apocrine breast lesions is based on the postulate that invasive apocrine carcinomas evolve from epithelial cells in terminal duct lobular units (TDLUs) in a stepwise manner that involves apocrine metaplasia of normal breast epithelia, hyperplasia, atypia, and apocrine carcinoma in situ. First, we identify specific protein biomarkers for benign apocrine metaplasia and thereafter we search for biomarkers that are highly overexpressed by pure invasive apocrine carcinomas. Here we present studies in which we have used antibodies against components of a benign apocrine signature that includes 15-prostaglandin dehydrogenase (15-PGDH), a protein that is expressed by all benign apocrine lesions, and markers that are highly overexpressed by pure invasive apocrine carcinomas such as MRP14 (S100A9), psoriasin (S100A7), and p53 to identify precancerous lesions in sclerosing adenosis (SA) with apocrine metaplasia. The latter is a benign proliferative lesion of the breast that exhibits an increase in the size of the TDLUs and characterized by retained two-cell lining, and myoepithelial (ME) and stromal hyperplasia. SA with apocrine metaplasia, i.e. apocrine adenosis (AA), presents with a higher degree of atypical apocrine hyperplasia, and these lesions are believed to be precursors of apocrine carcinoma, in situ and invasive. Analysis of 24 selected SA samples with apocrine metaplasia revealed non-obligate putative apocrine precancerous lesions that displayed some, or in same cases all the three markers associated with pure invasive apocrine carcinomas. These studies also revealed p53 positive, non-apocrine putative precancerous lesions as well as novel phenotypes for ME and some luminal cells characterized by the expression of cytokeratin 15.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Fibrocystic Breast Disease/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Precancerous Conditions/metabolism , Adult , Aged, 80 and over , Breast Neoplasms/pathology , Female , Fibrocystic Breast Disease/pathology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Metaplasia/metabolism , Metaplasia/pathology , Middle Aged , Precancerous Conditions/pathology
8.
Mol Oncol ; 1(2): 181-95, 2007 Sep.
Article in English | MEDLINE | ID: mdl-19383294

ABSTRACT

Transcript profiling of 27 normal colon mucosas and 258 adenocarcinomas showed Keratin23 to be increased in 78% microsatellite-stable tumors, while microsatellite-instable tumors showed low transcript levels, comparable to normal mucosas. Immunohistochemical analyses demonstrated that 88% of microsatellite-instable tumors were negative for Keratin23 protein, while 70% of MSS tumors and metastases derived from MSS-tumors showed high Keratin23 levels. Immunofluorescence analysis localized Keratin23 in the Golgi-apparatus. Golgi accumulation was unique for gastrointestinal adenocarcinomas. Immunoprecipitation and 2D-blot analysis revealed Keratin23 to be a 46.8 kDa phosphoprotein. Keratin23 impaired the proliferation of human colon cancer cells significantly, leading to cell death in microsatellite-instable but not microsatellite-stable cell lines, while COS7 cells experienced multiple nuclei and apoptosis. Keratin23 expression correlated significantly with transcription factor CEBPB. In conclusion, Keratin23 expression is a novel and important difference between microsatellite-stable and microsatellite-instable colon cancers.


Subject(s)
Adenocarcinoma/metabolism , Cell Proliferation , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Keratins, Type I/biosynthesis , Microsatellite Instability , Neoplasm Proteins/biosynthesis , Phosphoproteins/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , COS Cells , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Survival/genetics , Chlorocebus aethiops , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Gene Expression Profiling , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Keratins, Type I/genetics , Male , Transcription, Genetic/genetics , Tumor Cells, Cultured
9.
Mol Oncol ; 1(3): 321-49, 2007 Dec.
Article in English | MEDLINE | ID: mdl-19383306

ABSTRACT

Recently, we presented evidence--based on the analysis of benign hyperproliferative lesions of the breast--for the presence of cells that express the stem cell marker cytokeratin (CK) 15 in combination with CK19, a protein widely expressed by mammary epithelial cells. Here we report the finding of a subset of breast carcinomas characterized by expression of CK15. CK15 expressing tumors constituted 5% (6 out of 120; 4 of ductal type and 2 of lobular type) of the high-risk breast carcinomas examined by gel-based proteomics and immunohistochemistry. Five out of the six CK15+ carcinomas were CK15+/CK19-. The remaining tumor was mainly composed of cells expressing both CK15 and CK19 (CK15+/CK19+), but it also contained invasive areas with cells expressing only one of these makers (CK15+/CK19- and CK15-/CK19+ cells). To address the relationship between putative luminal progenitor/amplified CK15+ cells and malignant disease, and to determine whether cells/lesions lose expression of CK15 as a result of tumour initiation and/or progression, we searched among our sample set for carcinomas in which invasive tumor areas co-existed with non-malignant cells and hyperproliferative and known pre-malignant lesions. Only one such tumour was found (T71), a CK15-/CK19+/p53+ carcinoma that contained p53 negative non-malignant epithelial cells exhibiting a variety of, CK15/CK19 cellular phenotypes (CK15+/CK19+; CK15+/CK19-; CK15-/CK19+; CK15-/CK19-), often associated with simple columnar cells. Single layers of epithelial cells exhibiting all four CK15/CK19 phenotypes were observed contiguous to areas of atypical ductal hyperplasia that contained p53 positive cells that lost CK15 expression (CK15-/CK19+) and had a very similar phenotype to those of the neighboring ductal carcinoma in situ (DCIS) and invasive cells. The undifferentiated CK15+/CK19+ cells, which had the phenotype CK15+/CK19+/CK14+/CK8+ and -/ER-/PgR-/AR-/CD44+ (weak)/CK17-/p63-/vimentin+/Ki67-/Bcl-2+ (weak)/GATA-3-/p53-, most likely correspond to lineage-restricted luminal progenitor cells able to generate the other more differentiated CK15/CK19 cellular phenotypes, thus giving rise to the daunting intratumour heterogeneity displayed by carcinoma T71. Cells with a very similar phenotype to the CK15+/CK19+ progenitor cells were observed in a juvenile fibroadenoma as well as in the large collecting ducts of the breast. The latter, however, expressed in addition CK14 and had a phenotype (CK15+/CK19+/CK14+/CK8+ (weak)/ER-/PgR-/AR-/CD44+ (weak)/CK17-/p63-/vimentin-/Ki67-/Bcl-2+/GATA-3-/p53-) that resembled that of the putative normal adult breast stem cells as inferred from published data. Further molecular characterization of these progenitor cells as well as unraveling of the signaling pathways that regulate their growth and differentiation may prove invaluable for developing novel therapeutic strategies that target cancer at an early stage.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Keratin-15/metabolism , Precancerous Conditions/pathology , Stem Cells , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Female , Fluorescent Antibody Technique, Direct , Humans , Immunohistochemistry , Keratin-15/genetics , Middle Aged , Neoplasm Invasiveness , Precancerous Conditions/chemistry , Stem Cells/chemistry , Stem Cells/metabolism , Stem Cells/pathology
10.
FEBS Lett ; 580(12): 2935-44, 2006 May 22.
Article in English | MEDLINE | ID: mdl-16631754

ABSTRACT

Breast cancer is a heterogeneous disease that encompasses a wide range of histopathological types including: invasive ductal carcinoma, lobular carcinoma, medullary carcinoma, mucinous carcinoma, tubular carcinoma, and apocrine carcinoma among others. Pure apocrine carcinomas represent about 0.5% of all invasive breast cancers according to the Danish Breast Cancer Cooperative Group Registry, and despite the fact that they are morphologically distinct from other breast lesions, there are at present no standard molecular criteria available for their diagnosis. In addition, the relationship between benign apocrine changes and breast carcinoma is unclear and has been a matter of discussion for many years. Recent proteome expression profiling studies of breast apocrine macrocysts, normal breast tissue, and breast tumours have identified specific apocrine biomarkers [15-hydroxyprostaglandin dehydrogenase (15-PGDH) and hydroxymethylglutaryl coenzyme A reductase (HMG-CoA reductase)] present in early and advanced apocrine lesions. These biomarkers in combination with proteins found to be characteristically upregulated in pure apocrine carcinomas (psoriasin, S100A9, and p53) provide a protein expression signature distinctive for benign apocrine metaplasias and apocrine cystic lesions. These studies have also presented compelling evidence for a direct link, through the expression of the prostaglandin degrading enzyme 15-PGDH, between early apocrine lesions and pure apocrine carcinomas. Moreover, specific antibodies against the components of the expression signature have identified precursor lesions in the linear histological progression to apocrine carcinoma. Finally, the identification of proteins that characterize the early stages of mammary apocrine differentiation such as 15-PGDH, HMG-CoA reductase, and cyclooxygenase 2 (COX-2) has opened a window of opportunity for pharmacological intervention, not only in a therapeutic manner but also in a chemopreventive setting. Here we review published and recent results in the context of the current state of research on breast apocrine cancer.


Subject(s)
Apocrine Glands/pathology , Breast Neoplasms/pathology , Neoplasm Proteins/metabolism , Apocrine Glands/metabolism , Breast Neoplasms/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans
11.
Mol Cell Proteomics ; 5(3): 462-83, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16316978

ABSTRACT

Up to one-third of women aged 30-50 years have cysts in their breasts and are presumed to be at increased risk of developing breast cancer. Here we present an extensive proteomic and immunohistochemistry (IHC) study of breast apocrine cystic lesions aimed at generating specific biomarkers and elucidating the relationship, if existent, of apocrine cysts with cancer phenotype. To this end we compared the expression profiles of apocrine macrocysts obtained from mastectomies from high risk cancer patients with those of cancerous and non-malignant mammary tissue biopsies collected from the same patients. We identified two biomarkers, 15-hydroxyprostaglandin dehydrogenase and 3-hydroxymethylglutaryl-CoA reductase, that were expressed specifically by apocrine type I cysts as well as by apocrine metaplastic cells in type II microcysts, terminal ducts, and intraductal papillary lesions. No expression of these markers was observed in non-malignant terminal ductal lobular units, type II flat cysts, stroma cells, or fat tissue as judged by IHC analysis of matched non-malignant tissue samples collected from 93 high risk patients enrolled in our cancer program. IHC analysis of the corresponding 93 primary tumors indicated that most apocrine changes have little intrinsic malignant potential, although some may progress to invasive apocrine cancer. None of the apocrine lesions examined, however, seemed to be a precursor of invasive ductal carcinomas, which accounted for 81% of the tumors analyzed. Our studies also provided some insight into the origin, development, and enlargement of apocrine cysts in mammary tissue. The successful identification of differentially expressed proteins that characterize specific steps in the progression from early benign lesions to apocrine cancer opens a window of opportunity for designing and testing new approaches for pharmacological intervention, not only in a therapeutic setting but also for chemoprevention, to inhibit cyst development as both 15-hydroxyprostaglandin dehydrogenase and 3-hydroxymethylglutaryl-CoA reductase are currently being targeted for chemoprevention strategies in various malignancies.


Subject(s)
Apocrine Glands/pathology , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Breast/pathology , Cysts/pathology , Adult , Aged , Aged, 80 and over , Apocrine Glands/metabolism , Breast/metabolism , Breast Neoplasms/metabolism , Cyst Fluid/chemistry , Cysts/metabolism , Cytokines/metabolism , Female , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/chemistry , Neoplasm Staging , Patient Selection , Phenotype , Proteome/analysis , Proteome/chemistry , Reproducibility of Results
12.
Mol Cell Proteomics ; 4(4): 492-522, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15695426

ABSTRACT

It has become clear that growth and progression of breast tumor cells not only depend on their malignant potential but also on factors present in the tumor microenvironment. Of the cell types that constitute the mammary stroma, the adipocytes are perhaps the least well studied despite the fact that they represent one of the most prominent cell types surrounding the breast tumor cells. There is compelling evidence demonstrating a role for the mammary fat pad in mammary gland development, and some studies have revealed the ability of fat tissue to augment the growth and ability to metastasize of mammary carcinoma cells. Very little is known, however, about which factors adipocytes produce that may orchestrate these actions and how this may come about. In an effort to shed some light on these questions, we present here a detailed proteomic analysis, using two-dimensional gel-based technology, mass spectrometry, immunoblotting, and antibody arrays, of adipose cells and interstitial fluid of fresh fat tissue samples collected from sites topologically distant from the tumors of high risk breast cancer patients that underwent mastectomy and that were not treated prior to surgery. A total of 359 unique proteins were identified, including numerous signaling molecules, hormones, cytokines, and growth factors, involved in a variety of biological processes such as signal transduction and cell communication; energy metabolism; protein metabolism; cell growth and/or maintenance; immune response; transport; regulation of nucleobase, nucleoside, and nucleic acid metabolism; and apoptosis. Apart from providing a comprehensive overview of the mammary fat proteome and its interstitial fluid, the results offer some insight as to the role of adipocytes in the breast tumor microenvironment and provide a first glance of their molecular cellular circuitry. In addition, the results open new possibilities to the study of obesity, which has a strong association with type 2 diabetes, hypertension, and coronary heart disease.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/pathology , Intracellular Fluid/metabolism , Mammary Glands, Human/cytology , Risk , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Antibodies/immunology , Blotting, Western , Cell Extracts , Electrophoresis, Gel, Two-Dimensional , Female , Fluorescent Antibody Technique, Indirect , Humans , Immunohistochemistry , Mammary Glands, Human/surgery , Mass Spectrometry , Mastectomy , Octoxynol/pharmacology , Oligonucleotide Array Sequence Analysis , Peptide Mapping , Proteome/analysis , Proteomics/methods
13.
Mol Cell Proteomics ; 4(4): 534-44, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15671042

ABSTRACT

We initiated the present study to identify new genes associated with colorectal cancer. In a previously published microarray study an EST (W80763), later identified as the gene hFKBP10 (NM_021939), was found to be strongly expressed in tumors while absent in the normal mucosa. Here we describe this gene hFKBP10 together with its encoded protein hFKBP65 as a novel marker associated with colorectal cancer. Analysis of 31 colorectal adenocarcinomas and 14 normal colorectal mucosa by RealTime PCR for hFKBP10 showed a significant up-regulation in tumors, when compared with normal mucosa. Immunohistochemical analysis of 26 adenocarcinomas and matching normal mucosa, as well as benign hyperplastic polyps and adenomas, using a monoclonal anti-hFKBP65 antibody, showed that the protein was not present in normal colorectal epithelial cells, but strongly expressed in the tumor cells of colorectal cancer. The protein was also expressed in fibroblasts of both normal mucosa and tumor tissue. Western blot analysis of matched tumors and normal mucosa supported the finding of increased hFKBP65 expression in tumors compared with normal mucosa, in addition to identifying the molecular mass of hFKBP65 to approximately 72 kDa. Cellular localization and glycosylation studies revealed the hFKBP65 protein to be localized in the endoplasmic reticulum, and to be N-glycosylated. In conclusion, the protein hFKBP65 is associated with colorectal cancer, and we hypothesize the protein to be involved in fibroblast and transformed epithelial cell-specific protein synthesis in the endoplasmic reticulum.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Tacrolimus Binding Proteins/metabolism , Adenocarcinoma/pathology , Adenoma/pathology , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites , Blotting, Western , COS Cells , Chlorocebus aethiops , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression Regulation, Neoplastic , Humans , Hyperplasia , Immunohistochemistry , Intestinal Mucosa/cytology , Isoelectric Point , Male , Microscopy, Fluorescence , Middle Aged , Molecular Sequence Data , Molecular Weight , Polymerase Chain Reaction , Protein Binding , Tacrolimus Binding Proteins/analysis , Tacrolimus Binding Proteins/chemistry , Transfection
14.
FEBS J ; 272(1): 2-15, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15634327

ABSTRACT

Discovery-driven translational research in breast cancer is moving steadily from the study of cell lines to the analysis of clinically relevant samples that, together with the ever increasing number of novel and powerful technologies available within genomics, proteomics and functional genomics, promise to have a major impact on the way breast cancer will be diagnosed, treated and monitored in the future. Here we present a brief report on long-term ongoing strategies at the Danish Centre for Translational Breast Cancer Research to search for markers for early detection and targets for therapeutic intervention, to identify signalling pathways affected in individual tumours, as well as to integrate multiplatform 'omic' data sets collected from tissue samples obtained from individual patients. The ultimate goal of this initiative is to coalesce knowledge-based complementary procedures into a systems biology approach to fight breast cancer.


Subject(s)
Breast Neoplasms/genetics , Protein Biosynthesis , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Humans , Research
15.
Pharmacogenomics ; 5(4): 381-94, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15165174

ABSTRACT

Detecting bladder cancer at an early stage and predicting how a tumor will behave and act in response to therapy, as well as the identification of new targets for therapeutic intervention, are among the main areas of research that will benefit from the current explosion in the number of powerful technologies emerging within proteomics. The purpose of this article is to briefly review what has been achieved to date using proteomic technologies and to bring forward novel strategies - based on the analysis of clinically relevant samples - that promise to accelerate the translation of basic discoveries into the daily clinical practice.


Subject(s)
Proteomics/methods , Urinary Bladder Neoplasms/genetics , Animals , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Humans , Proteomics/trends , Urinary Bladder Neoplasms/drug therapy
16.
Mol Cell Proteomics ; 3(4): 327-44, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14754989

ABSTRACT

Clinical cancer proteomics aims at the identification of markers for early detection and predictive purposes, as well as to provide novel targets for drug discovery and therapeutic intervention. Proteomics-based analysis of traditional sources of biomarkers, such as serum, plasma, or tissue lyzates, has resulted in a wealth of information and the finding of several potential tumor biomarkers. However, many of these markers have shown limited usefulness in a clinical setting, underscoring the need for new clinically relevant sources. Here we present a novel and highly promising source of biomarkers, the tumor interstitial fluid (TIF) that perfuses the breast tumor microenvironment. We collected TIFs from small pieces of freshly dissected invasive breast carcinomas and analyzed them by two-dimensional polyacrylamide gel electrophoresis in combination with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, Western immunoblotting, as well as by cytokine-specific antibody arrays. This approach provided for the first time a snapshot of the protein components of the TIF, which we show consists of more than one thousand proteins--either secreted, shed by membrane vesicles, or externalized due to cell death--produced by the complex network of cell types that make up the tumor microenvironment. So far, we have identified 267 primary translation products including, but not limited to, proteins involved in cell proliferation, invasion, angiogenesis, metastasis, inflammation, protein synthesis, energy metabolism, oxidative stress, the actin cytoskeleton assembly, protein folding, and transport. As expected, the TIF contained several classical serum proteins. Considering that the protein composition of the TIF reflects the physiological and pathological state of the tissue, it should provide a new and potentially rich resource for diagnostic biomarker discovery and for identifying more selective targets for therapeutic intervention.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/diagnosis , Extracellular Fluid/chemistry , Proteome , Proteomics , Adult , Aged , Aged, 80 and over , Blotting, Western , Breast Neoplasms/therapy , Electrophoresis, Gel, Two-Dimensional/methods , Female , Gene Expression Profiling , Humans , Middle Aged , Neoplasm Invasiveness/pathology , Peptide Fragments/chemistry , Peptide Mapping , Perfusion , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Mol Cell Proteomics ; 2(6): 369-77, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12832461

ABSTRACT

The application of state-of-the-art proteomics and functional genomics technologies to the study of cancer is rapidly shifting toward the analysis of clinically relevant samples derived from patients, as the ultimate aim of translational research is to bring basic discoveries closer to the bedside. Here we describe the essence of a long-term initiative undertaken by The Danish Centre for Translational Breast Cancer Research and currently underway for cancer biomarker discovery using fresh tissue biopsies and bio-fluids. The Centre is a virtual hub that brings together scientists working in various areas of basic cancer research such as cell cycle control, invasion and micro-environmental alterations, apoptosis, cell signaling, and immunology, with clinicians (oncologists, surgeons), pathologists, and epidemiologists, with the aim of understanding the molecular mechanisms underlying breast cancer progression and ultimately of improving patient survival and quality of life. The unifying concept behind our approach is the use of various experimental paradigms for the prospective analysis of clinically relevant samples obtained from the same patient, along with the systematic integration of the biological and clinical data.


Subject(s)
Academies and Institutes , Biomarkers, Tumor , Biomedical Research/trends , Breast Neoplasms/metabolism , Genomics , Proteomics , Biopsy , Body Fluids/chemistry , Breast Neoplasms/classification , Breast Neoplasms/genetics , Denmark , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Humans , Mammary Glands, Human/pathology , Pilot Projects , Specimen Handling
SELECTION OF CITATIONS
SEARCH DETAIL
...