Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Immunother Cancer ; 12(4)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38604809

ABSTRACT

BACKGROUND: Combining cytotoxic chemotherapy or novel anticancer drugs with T-cell modulators holds great promise in treating advanced cancers. However, the response varies depending on the tumor immune microenvironment (TIME). Therefore, there is a clear need for pharmacologically tractable models of the TIME to dissect its influence on mono- and combination treatment response at the individual level. METHODS: Here we establish a patient-derived explant culture (PDEC) model of breast cancer, which retains the immune contexture of the primary tumor, recapitulating cytokine profiles and CD8+T cell cytotoxic activity. RESULTS: We explored the immunomodulatory action of a synthetic lethal BCL2 inhibitor venetoclax+metformin drug combination ex vivo, discovering metformin cannot overcome the lymphocyte-depleting action of venetoclax. Instead, metformin promotes dendritic cell maturation through inhibition of mitochondrial complex I, increasing their capacity to co-stimulate CD4+T cells and thus facilitating antitumor immunity. CONCLUSIONS: Our results establish PDECs as a feasible model to identify immunomodulatory functions of anticancer drugs in the context of patient-specific TIME.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Bridged Bicyclo Compounds, Heterocyclic , Metformin , Sulfonamides , Humans , Female , Electron Transport Complex I/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dendritic Cells , Metformin/pharmacology , Metformin/therapeutic use , Tumor Microenvironment
2.
Stem Cell Res ; 74: 103301, 2024 02.
Article in English | MEDLINE | ID: mdl-38176368

ABSTRACT

Characterized human induced pluripotent stem cell lines are important for basic research. Here, we report the establishment of three isogenic human induced pluripotent stem cell (hiPSC) lines generated from normal neonate male skin fibroblasts. Pluripotency was induced using the integration free Sendai virus reprogramming method. The pluripotency, identity, quality, and safety of the lines were confirmed to establish characterized human induced pluripotent stem cell lines to be used as normal control cell lines in future studies.


Subject(s)
Induced Pluripotent Stem Cells , Infant, Newborn , Humans , Male , Induced Pluripotent Stem Cells/metabolism , Cellular Reprogramming , Cell Differentiation , Cell Line , Fibroblasts/metabolism
3.
Nat Commun ; 12(1): 3904, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162871

ABSTRACT

Due to its dynamic nature, the evolution of cancer cell-extracellular matrix (ECM) crosstalk, critically affecting metastasis and treatment resistance, remains elusive. Our results show that platinum-chemotherapy itself enhances resistance by progressively changing the cancer cell-intrinsic adhesion signaling and cell-surrounding ECM. Examining ovarian high-grade serous carcinoma (HGSC) transcriptome and histology, we describe the fibrotic ECM heterogeneity at primary tumors and distinct metastatic sites, prior and after chemotherapy. Using cell models from systematic ECM screen to collagen-based 2D and 3D cultures, we demonstrate that both specific ECM substrates and stiffness increase resistance to platinum-mediated, apoptosis-inducing DNA damage via FAK and ß1 integrin-pMLC-YAP signaling. Among such substrates around metastatic HGSCs, COL6 was upregulated by chemotherapy and enhanced the resistance of relapse, but not treatment-naïve, HGSC organoids. These results identify matrix adhesion as an adaptive response, driving HGSC aggressiveness via co-evolving ECM composition and sensing, suggesting stromal and tumor strategies for ECM pathway targeting.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , Collagen/genetics , Collagen/metabolism , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Evolution, Molecular , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Humans , Kaplan-Meier Estimate , Neoplasm Recurrence, Local , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
4.
Gastric Cancer ; 24(6): 1254-1263, 2021 11.
Article in English | MEDLINE | ID: mdl-34115237

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the third most common cause of cancer death. Intestinal type GC is a molecularly diverse disease. Formins control cytoskeletal processes and have been implicated in the progression of many cancers. Their clinical significance in GC remains unclear. Here, we characterize the expression of formin proteins FHOD1 and FMNL1 in intestinal GC tissue samples and investigate their association with clinical parameters, GC molecular subtypes and intratumoral T lymphocytes. METHODS: The prognostic significance of FHOD1 and FMNL1 mRNA expression was studied with Kaplan-Meier analyses in an online database. The expression of FHOD1 and FMNL1 proteins was characterized in GC cells, and in non-neoplastic and malignant tissues utilizing tumor microarrays of intestinal GC representing different molecular subtypes. FHOD1 and FMNL1 expression was correlated with clinical parameters, molecular features and T lymphocyte infiltration. Immunohistochemical expression of neither formin correlated with survival. RESULTS: Kaplan-Meier analysis associated high FHOD1 and FMNL1 mRNA expression with reduced overall survival (OS). Characterization of FHOD1 and FMNL1 in GC cells showed cytoplasmic expression along the actin filaments. Similar pattern was recapitulated in GC tissue samples. Elevated FMNL1 was associated with larger tumor size and higher disease stage. Downregulation of FHOD1 associated with TP53-mutated GC tumors. Tumor cell FHOD1 expression strongly correlated with high numbers of tumor-infiltrating CD8 + lymphocytes. CONCLUSIONS: FHOD1 and FMNL1 proteins are expressed in the tumor cells of intestinal GC and significantly associate with clinical parameters without direct prognostic significance. FHOD1 correlates with high intratumoral CD8 + T lymphocyte infiltration in this cohort.


Subject(s)
Lymphocytes, Tumor-Infiltrating/metabolism , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Fetal Proteins/metabolism , Finland , Formins/metabolism , Humans , Male , Middle Aged , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Analysis
5.
Sci Rep ; 11(1): 8856, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893375

ABSTRACT

Erythropoietin producing hepatocellular (Eph) receptors and their membrane-bound ligands ephrins are variably expressed in epithelial cancers, with context-dependent implications to both tumor-promoting and -suppressive processes in ways that remain incompletely understood. Using ovarian cancer tissue microarrays and longitudinally collected patient cells, we show here that ephrinA5/EFNA5 is specifically overexpressed in the most aggressive high-grade serous carcinoma (HGSC) subtype, and increased in the HGSC cells upon disease progression. Among all the eight ephrin genes, high EFNA5 expression was most strongly associated with poor overall survival in HGSC patients from multiple independent datasets. In contrast, high EFNA3 predicted improved overall and progression-free survival in The Cancer Genome Atlas HGSC dataset, as expected for a canonical inducer of tumor-suppressive Eph receptor tyrosine kinase signaling. While depletion of either EFNA5 or the more extensively studied, canonically acting EFNA1 in HGSC cells increased the oncogenic EphA2-S897 phosphorylation, EFNA5 depletion left unaltered, or even increased the ligand-dependent EphA2-Y588 phosphorylation. Moreover, treatment with recombinant ephrinA5 led to limited EphA2 tyrosine phosphorylation, internalization and degradation compared to ephrinA1. Altogether, our results suggest a unique function for ephrinA5 in Eph-ephrin signaling and highlight the clinical potential of ephrinA5 as a cell surface biomarker in the most aggressive HGSCs.


Subject(s)
Ephrin-A5/metabolism , Neoplasm Recurrence, Local , Ovarian Neoplasms/pathology , Receptor, EphA2/metabolism , Up-Regulation , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Ovarian Neoplasms/metabolism , Signal Transduction , Survival Analysis
6.
Virchows Arch ; 478(4): 707-717, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32954467

ABSTRACT

While host immune response is likely to be important for the prognosis of gastric cancer patients, detailed information on the T lymphocyte infiltration in different gastric cancer subtypes is lacking. Here, we studied the presence of CD3, CD8, and FOXP3 (Forkhead box p3) expressing T lymphocytes in a retrospective cohort of 190 intestinal gastric and gastroesophageal adenocarcinomas. The cancers represented four distinct molecular subtypes: Epstein-Barr virus-positive (EBV+), mismatch-repair-deficient (MMR-D), aberrant TP53, and the "other" subtype. The absolute numbers of CD3+, CD8+, and FOXP3+ T lymphocytes were analyzed in relation with these molecular subtypes and selected clinicopathological parameters. Overall, there was a large variation in the amount of infiltrating T lymphocyte in all molecular subtypes. Among the subtypes, EBV+ cancers differed from the other subtypes in increased lymphocyte infiltration and high CD8+/FOXP3+ ratio. While the TP53 aberrant subtype did not differ in the absolute amount of T lymphocyte, the ratio of CD8+/FOXP3+ and CD3+/FOXP3+ cells was highest in this subtype, possibly reflecting immunosuppression associated with genomic instability. Increased CD3+ and CD8+ T lymphocyte infiltrates were associated with better survival, and remained as independent prognostic factors in a multivariate analysis. This study is the first to investigate lymphocytic infiltration within four molecular subtypes of intestinal-type gastric cancer in a European cohort. The results provide an important addition to the current knowledge of T lymphocyte-dependent immune response in gastric cancer and its prognostic significance.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Follow-Up Studies , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Analysis , Tissue Array Analysis
7.
BMC Cancer ; 20(1): 710, 2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32727404

ABSTRACT

BACKGROUND: The prognosis of glioblastoma remains poor, related to its diffuse spread within the brain. There is an ongoing search for molecular regulators of this particularly invasive behavior. One approach is to look for actin regulating proteins that might be targeted by future anti-cancer therapy. The formin family of proteins orchestrates rearrangement of the actin cytoskeleton in multiple cellular processes. Recently, the formin proteins mDia1 and mDia2 were shown to be expressed in glioblastoma in vitro, and their function could be modified by small molecule agonists. This finding implies that the formins could be future therapeutic targets in glioblastoma. METHODS: In cell studies, we investigated the changes in expression of the 15 human formins in primary glioblastoma cells and commercially available glioblastoma cell lines during differentiation from spheroids to migrating cells using transcriptomic analysis and qRT-PCR. siRNA mediated knockdown of selected formins was performed to investigate whether their expression affects glioblastoma migration. Using immunohistochemistry, we studied the expression of two formins, FHOD1 and INF2, in tissue samples from 93 IDH-wildtype glioblastomas. Associated clinicopathological parameters and follow-up data were utilized to test whether formin expression correlates with survival or has prognostic value. RESULTS: We found that multiple formins were upregulated during migration. Knockdown of individual formins mDia1, mDia2, FHOD1 and INF2 significantly reduced migration in most studied cell lines. Among the studied formins, knockdown of INF2 generated the greatest reduction in motility in vitro. Using immunohistochemistry, we demonstrated expression of formin proteins FHOD1 and INF2 in glioblastoma tissues. Importantly, we found that moderate/high expression of INF2 was associated with significantly impaired prognosis. CONCLUSIONS: Formins FHOD1 and INF2 participate in glioblastoma cell migration. Moderate/high expression of INF2 in glioblastoma tissue is associated with worse outcome. Taken together, our in vitro and tissue studies suggest a pivotal role for INF2 in glioblastoma. When specific inhibiting compounds become available, INF2 could be a target in the search for novel glioblastoma therapies.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement , Fetal Proteins/metabolism , Formins/metabolism , Glioblastoma/metabolism , Actins/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Fetal Proteins/genetics , Formins/genetics , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Up-Regulation
8.
J Pathol ; 250(2): 159-169, 2020 02.
Article in English | MEDLINE | ID: mdl-31595974

ABSTRACT

Poor chemotherapy response remains a major treatment challenge for high-grade serous ovarian cancer (HGSC). Cancer stem cells are the major contributors to relapse and treatment failure as they can survive conventional therapy. Our objectives were to characterise stemness features in primary patient-derived cell lines, correlate stemness markers with clinical outcome and test the response of our cells to both conventional and exploratory drugs. Tissue and ascites samples, treatment-naive and/or after neoadjuvant chemotherapy, were prospectively collected. Primary cancer cells, cultured under conditions favouring either adherent or spheroid growth, were tested for stemness markers; the same markers were analysed in tissue and correlated with chemotherapy response and survival. Drug sensitivity and resistance testing was performed with 306 oncology compounds. Spheroid growth condition HGSC cells showed increased stemness marker expression (including aldehyde dehydrogenase isoform I; ALDH1A1) as compared with adherent growth condition cells, and increased resistance to platinum and taxane. A set of eight stemness markers separated treatment-naive tumours into two clusters and identified a distinct subgroup of HGSC with enriched stemness features. Expression of ALDH1A1, but not most other stemness markers, was increased after neoadjuvant chemotherapy and its expression in treatment-naive tumours correlated with chemoresistance and reduced survival. In drug sensitivity and resistance testing, five compounds, including two PI3K-mTOR inhibitors, demonstrated significant activity in both cell culture conditions. Thirteen compounds, including EGFR, PI3K-mTOR and aurora kinase inhibitors, were more toxic to spheroid cells than adherent cells. Our results identify stemness markers in HGSC that are associated with a decreased response to conventional chemotherapy and reduced survival if expressed by treatment-naive tumours. EGFR, mTOR-PI3K and aurora kinase inhibitors are candidates for targeting this cell population. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Antineoplastic Agents/pharmacology , Cystadenocarcinoma, Serous/pathology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Retinal Dehydrogenase/metabolism , Aurora Kinases/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant/methods , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/methods , ErbB Receptors/antagonists & inhibitors , Female , Humans , Molecular Targeted Therapy/methods , Neoplasm Grading , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Prognosis , Spheroids, Cellular/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured/drug effects
9.
Sci Rep ; 9(1): 10038, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296879

ABSTRACT

The analysis of extracellular vesicles (EVs) typically requires tedious and time-consuming isolation process from bio-fluids. We developed a nanoparticle-based time resolved fluorescence immunoassay (NP-TRFIA) that uses biotinylated antibodies against the proteins of tetraspanin family and tumor-associated antigens for capturing EVs from urine samples and cell culture supernatants without the need for isolation. The captured-EVs were detected either with Eu3+-chelate or Eu3+-doped nanoparticle-based labels conjugated either to antibodies against the tetraspanins or lectins targeting the glycan moieties on EVs surface. The NP-TRFIA demonstrated specific capturing and detection of EVs by antibodies and lectins. Lectin-nanoparticle based assays showed 2-10 fold higher signal-to-background ratio compared with lectin-chelate assays. The nanoparticle assay concept allowed surface glycosylation profiling of the urine derived-EVs with lectins. It was also applied to establish an assay showing differential expression of tumor-associated proteins on more aggressive (higher ITGA3 on DU145- and PC3-EVs) compared to less aggressive (higher EpCAM on LNCaP-EVs) PCa- cell lines derived-EVs. This NP-TRFIA can be used as a simple tool for analysis and characterization of EVs in urine and cell culture supernatants. Such approach could be useful in identification of disease-specific markers on the surface of patient-derived urinary EVs.


Subject(s)
Extracellular Vesicles/metabolism , Glycoproteins/analysis , Immunoassay/methods , Nanoparticles/chemistry , Tetraspanins/analysis , Adult , Antibodies/immunology , Biomarkers/analysis , Cell Line, Tumor , Female , Glycoproteins/immunology , Glycosylation , Humans , Lectins/metabolism , Male , Prostatic Neoplasms/diagnosis , Tetraspanins/immunology , Urine/chemistry
10.
Tumour Biol ; 40(3): 1010428318761494, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29542402

ABSTRACT

Expression of regenerating islet-derived protein 4 (REG4), a secretory protein involved in cell differentiation and proliferation, is upregulated in inflammatory bowel diseases and in many gastrointestinal malignancies. The prognostic significance of its expression in pancreatic ductal adenocarcinoma is unknown. Our aim was to investigate tumor tissue and serum REG4 expression in pancreatic ductal adenocarcinoma patients. We also evaluated as a control the diagnostic value of serum REG4 level in patients with chronic pancreatitis. Immunohistochemical expression of REG4 was evaluated in 154 surgical specimens and serum REG4 level in 130 samples from pancreatic ductal adenocarcinoma patients treated at Helsinki University Hospital, Finland, in 2000-2011. REG4 tissue and serum expression was assessed in relation to clinicopathological parameters and patient survival. A chronic pancreatitis control group comprised 34 patients who underwent pancreatic resection because of suspicion of malignancy. Significant survival differences were detectable in subgroups: in tumor stages IA-IIA, high serum REG4 level predicted worse survival (p=0.046). In patients with grade I tumor, positive tissue REG4 expression predicted better survival (p=0.006). In multivariate analysis, neither tissue nor serum REG4 expression was independent prognostic factors. Serum REG4 levels were higher in pancreatic ductal adenocarcinoma than in chronic pancreatitis (p=0.002), with diagnostic sensitivity of 45% and specificity of 91%. In logistic regression analysis, a multivariate model with REG4, CA19-9, and age provided sensitivity of 82% and specificity of 79%. REG4 tissue expression is a prognostic marker in subgroups of pancreatic ductal adenocarcinoma patients. Serum REG4 level might be useful in differential diagnosis between pancreatic ductal adenocarcinoma and chronic pancreatitis.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Pancreatic Ductal/blood , Pancreatic Neoplasms/blood , Pancreatitis-Associated Proteins/blood , Adult , Aged , Biomarkers, Tumor/biosynthesis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/surgery , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/surgery , Pancreatitis, Chronic/blood , Pancreatitis, Chronic/diagnosis , Pancreatitis, Chronic/metabolism , Pancreatitis-Associated Proteins/biosynthesis , Prognosis , Proportional Hazards Models
11.
J Pathol Clin Res ; 3(2): 123-138, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28451461

ABSTRACT

Breast cancer is the leading cause of cancer-related deaths in women due to distinct cancer subtypes associated with early recurrence and aggressive metastatic progression. High lipoprotein-associated phospholipase A2 (PLA2G7) expression has previously been associated with aggressive disease and metastasis in prostate cancer. Here, we explore the expression pattern and functional role of PLA2G7 in breast cancer. First, a bioinformatic analysis of genome-wide gene expression data from 970 breast samples was carried out to evaluate the expression pattern of PLA2G7 mRNA in breast cancer. Second, the expression profile of PLA2G7 was studied in 1042 breast cancer samples including 89 matched lymph node metastasis samples using immunohistochemistry. Third, the effect of PLA2G7 silencing on genome-wide gene expression profile was studied and validated in cultured breast cancer cells expressing PLA2G7 at high level. Last, the expression pattern of PLA2G7 mRNA was investigated in 24 nonmalignant tissue samples and 65 primary and 7 metastatic tumour samples derived from various organs using qRT-PCR. The results from clinical breast cancer samples indicated that PLA2G7 is overexpressed in a subset of breast cancer samples compared to its expression in benign breast tissue samples and that high PLA2G7 expression associated with hormone receptor negativity as well as with poor prognosis in a subset of breast cancer samples. In vitro functional studies highlighted the putative role of PLA2G7 in the regulation of epithelial-mesenchymal transition (EMT)-related signalling pathways, vimentin and E-cadherin protein expression as well as cell migration in cultured breast cancer cells. Furthermore, supporting the findings in breast and prostate cancer, high PLA2G7 mRNA expression was associated with metastatic cancer in four additional organs of origin. In conclusion, our results indicate that PLA2G7 is highly expressed in a subset of metastatic and aggressive breast cancers and in metastatic samples of various tissues of origin and promotes EMT and migration in cultured breast cancer cells.

12.
PLoS One ; 11(3): e0151590, 2016.
Article in English | MEDLINE | ID: mdl-26981633

ABSTRACT

Preoperative diagnostics of ovarian neoplasms rely on ultrasound imaging and the serum biomarkers CA125 and HE4. However, these markers may be elevated in non-neoplastic conditions and may fail to identify most non-serous epithelial cancer subtypes. The objective of this study was to identify histotype-specific serum biomarkers for mucinous ovarian cancer. The candidate genes with mucinous histotype specific expression profile were identified from publicly available gene-expression databases and further in silico data mining was performed utilizing the MediSapiens database. Candidate biomarker validation was done using qRT-PCR, western blotting and immunohistochemical staining of tumor tissue microarrays. The expression level of the candidate gene in serum was compared to the serum CA125 and HE4 levels in a patient cohort of prospectively collected advanced ovarian cancer. Database searches identified REG4 as a potential biomarker with specificity for the mucinous ovarian cancer subtype. The specific expression within epithelial ovarian tumors was further confirmed by mRNA analysis. Immunohistochemical staining of ovarian tumor tissue arrays showed distinctive cytoplasmic expression pattern only in mucinous carcinomas and suggested differential expression between benign and malignant mucinous neoplasms. Finally, an ELISA based serum biomarker assay demonstrated increased expression only in patients with mucinous ovarian cancer. This study identifies REG4 as a potential serum biomarker for histotype-specific detection of mucinous ovarian cancer and suggests serum REG4 measurement as a non-invasive diagnostic tool for postoperative follow-up of patients with mucinous ovarian cancer.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Lectins, C-Type/metabolism , Ovarian Neoplasms/metabolism , Adenocarcinoma, Mucinous/blood , Adenocarcinoma, Mucinous/pathology , Biomarkers, Tumor/metabolism , CA-125 Antigen/blood , Databases, Factual , Female , Humans , Lectins, C-Type/blood , Ovarian Neoplasms/blood , Ovarian Neoplasms/pathology , Pancreatitis-Associated Proteins , Proteins/metabolism , WAP Four-Disulfide Core Domain Protein 2
13.
PLoS One ; 8(9): e74923, 2013.
Article in English | MEDLINE | ID: mdl-24086398

ABSTRACT

Cancer cells can obtain their ability to invade and metastasise by undergoing epithelial-to-mesenchymal transition (EMT). Exploiting this mechanism of cellular plasticity, malignant cells can remodel their actin cytoskeleton and down-regulate proteins needed for cell-cell contacts. The mechanisms of cytoskeletal reorganisation resulting in mesenchymal morphology and increased invasive potential are poorly understood. Actin nucleating formins have been implicated as key players in EMT. Here, we analysed which formins are altered in squamous cell carcinoma related EMT. FHOD1, a poorly studied formin, appeared to be markedly upregulated upon EMT. In human tissues FHOD1 was primarily expressed in mesenchymal cells, with little expression in epithelia. However, specimens from oral squamous cell cancers demonstrated consistent FHOD1 upregulation in mesenchymally transformed cells at the invasive edge. This upregulation was confirmed in an oral squamous carcinoma model, where FHOD1 expression was markedly increased upon EMT in a PI3K signalling dependent manner. In the EMT cells FHOD1 contributed to the spindle-shaped morphology and mesenchymal F-actin organization. Furthermore, functional assays demonstrated that FHOD1 contributes to cell migration and invasion. Finally, FHOD1 depletion reduced the ability of EMT cancer cells to form invadopodia and to degrade extracellular matrix. Our results indicate that FHOD1 participates in cytoskeletal changes in EMT. In addition, we show that FHOD1 upregulation occurs during cancer cell EMT in vivo, which indicates that FHOD1 may contribute to tumour progression.


Subject(s)
Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Fetal Proteins/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Nuclear Proteins/metabolism , Up-Regulation/genetics , Aged , Cell Line, Tumor , Cell Shape , Endothelium/metabolism , Epithelial Cells/metabolism , Female , Fetal Proteins/genetics , Formins , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Humans , Mesoderm/pathology , Mouth Neoplasms/enzymology , Neoplasm Invasiveness , Neoplasms, Squamous Cell/enzymology , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/pathology , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Plasma Cells/metabolism , Proteolysis , RNA, Small Interfering/metabolism , Signal Transduction , Transcription, Genetic , Transcriptome/genetics
14.
Oncotarget ; 4(1): 48-63, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23295955

ABSTRACT

Vimentin is an intermediate filament protein, with a key role in the epithelial to mesenchymal transition as well as cell invasion, and it is often upregulated during cancer progression. However, relatively little is known about its regulation in cancer cells. Here, we performed an RNA interference screen followed by protein lysate microarray analysis in bone metastatic MDA-MB-231(SA) breast cancer cells to identify novel regulators of vimentin expression. Out of the 596 genes investigated, three novel vimentin regulators EPHB4, WIPF2 and MTHFD2 were identified. The reduced vimentin expression in response to EPHB4, WIPF2 and MTHFD2 silencing was observed at mRNA and protein levels. Bioinformatic analysis of gene expression data across cancers indicated overexpression of EPHB4 and MTHFD2 in breast cancer and high expression associated with poor clinical characteristics. Analysis of 96 cDNA samples derived from both normal and malignant human tissues suggested putative association with metastatic disease. MTHFD2 knockdown resulted in impaired cell migration and invasion into extracellular matrix as well as decreased the fraction of cells with a high CD44 expression, a marker of cancer stem cells. Furthermore, MTHFD2 expression was induced in response to TGF-ß stimulation in breast cancer cells. Our results show that MTHFD2 is overexpressed in breast cancer, associates with poor clinical characteristics and promotes cellular features connected with metastatic disease, thus implicating MTHFD2 as a potential drug target to block breast cancer cell migration and invasion.


Subject(s)
Aminohydrolases/genetics , Breast Neoplasms/genetics , Cell Movement/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , RNA Interference , Vimentin/genetics , Aminohydrolases/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Microfilament Proteins , Microscopy, Confocal , Multienzyme Complexes/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology , Vimentin/metabolism
15.
J Pathol ; 226(4): 674-86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22072156

ABSTRACT

Breast cancer is the most frequent cancer and the leading cause of cancer-related deaths in women worldwide. The prognosis of breast cancer is tightly correlated with the degree of spread beyond the primary tumour. Arachidonic acid (AA) and prostaglandin E(2) (PGE(2)) are known to regulate tumour metastasis enabling epithelial-mesenchymal transition (EMT). However, the detailed role of 15-hydroxyprostaglandin dehydrogenase (HPGD), the key enzyme degrading prostaglandin E(2) , remains unclear in breast cancer. Here, we show that HPGD mRNA is overexpressed in a subset of clinical breast cancers compared to normal breast tissue samples and that high HPGD mRNA expression associates with poor prognosis. Immunohistochemical staining of primary breast cancer and lymph node metastasis tissue samples confirmed high HPGD protein expression in 20% of the samples, as well as associated HPGD expression with aggressive characteristics, such as increased risk of disease relapse and shorter disease-free survival. Results from cultured cells indicated abundant HPGD expression in highly metastatic breast cancer cells, and impairment of HPGD expression using RNA interference led to a significant decrease in transforming growth factor-ß signalling, in cellular arachidonic acid levels as well as in cell migration. Furthermore, gene expression microarray analysis followed by quantitative RT-PCR validation showed that HPGD silencing decreased aryl hydrocarbon receptor signalling and induced mesenchymal-epithelial transition. In conclusion, our results indicate that HPGD is highly expressed in metastatic and aggressive breast cancer and promotes EMT and migration in breast cancer cells.


Subject(s)
Adenocarcinoma/secondary , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Hydroxyprostaglandin Dehydrogenases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Arachidonic Acid/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Adhesion , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Germany/epidemiology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Lymph Nodes/metabolism , Lymph Nodes/pathology , Prognosis , Survival Rate , Tissue Array Analysis , Wound Healing
16.
Oncotarget ; 2(12): 1176-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22202492

ABSTRACT

Prostate cancer is the second leading cause of cancer mortality in men in developed countries. Due to the heterogeneous nature of the disease, design of novel personalized treatments is required to achieve efficient therapeutic responses. We have recently identified phospholipase 2 group VII (PLA2G7) as a potential drug target especially in ERG oncogene positive prostate cancers. Here, the expression profile of PLA2G7 was studied in 1137 prostate cancer and 409 adjacent non-malignant prostate tissues using immunohistochemistry to validate its biomarker potential and putative association with disease progression. In order to reveal the molecular alterations induced by PLA2G7 impairment, lipidomic and gene expression profiling was performed in response to PLA2G7 silencing in cultured prostate cancer cells. Moreover, the antineoplastic effect of statins combined with PLA2G7 impairment was studied in prostate cancer cells to evaluate the potential of repositioning of in vivo compatible drugs developed for other indications towards anti-cancer purposes. The results indicated that PLA2G7 is a cancer-selective biomarker in 50 % of prostate cancers and associates with aggressive disease. The alterations induced by PLA2G7 silencing highlighted the potential of PLA2G7 inhibition as an anti-proliferative, pro-apoptotic and anti-migratorial therapeutic approach in prostate cancer. Moreover, the anti-proliferative effect of PLA2G7 silencing was potentiated by lipid-lowering statins in prostate cancer cells. Taken together, our results support the potential of PLA2G7 as a biomarker and a drug target in prostate cancer and present a rationale for combining PLA2G7 inhibition with the use of statins in prostate cancer management.


Subject(s)
Cell Movement , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Phospholipases A2/metabolism , Prostatic Neoplasms/enzymology , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Aldehyde Dehydrogenase/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Biomarkers, Tumor , Caspase 3/metabolism , Caspase 7/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lysophosphatidylcholines/metabolism , Male , Neoplasm Invasiveness/genetics , Phospholipases A2/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering
17.
Cancer Res ; 71(9): 3236-45, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21415164

ABSTRACT

Activation of lipid metabolism is an early event in carcinogenesis and a central hallmark of many cancers. However, the precise molecular composition of lipids in tumors remains generally poorly characterized. The aim of the present study was to analyze the global lipid profiles of breast cancer, integrate the results to protein expression, and validate the findings by functional experiments. Comprehensive lipidomics was conducted in 267 human breast tissues using ultraperformance liquid chromatography/ mass spectrometry. The products of de novo fatty acid synthesis incorporated into membrane phospholipids, such as palmitate-containing phosphatidylcholines, were increased in tumors as compared with normal breast tissues. These lipids were associated with cancer progression and patient survival, as their concentration was highest in estrogen receptor-negative and grade 3 tumors. In silico transcriptomics database was utilized in investigating the expression of lipid metabolism related genes in breast cancer, and on the basis of these results, the expression of specific proteins was studied by immunohistochemistry. Immunohistochemical analyses showed that several genes regulating lipid metabolism were highly expressed in clinical breast cancer samples and supported also the lipidomics results. Gene silencing experiments with seven genes [ACACA (acetyl-CoA carboxylase α), ELOVL1 (elongation of very long chain fatty acid-like 1), FASN (fatty acid synthase), INSIG1 (insulin-induced gene 1), SCAP (sterol regulatory element-binding protein cleavage-activating protein), SCD (stearoyl-CoA desaturase), and THRSP (thyroid hormone-responsive protein)] indicated that silencing of multiple lipid metabolism-regulating genes reduced the lipidomic profiles and viability of the breast cancer cells. Taken together, our results imply that phospholipids may have diagnostic potential as well as that modulation of their metabolism may provide therapeutic opportunities in breast cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , Lipid Metabolism/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Disease Progression , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Gene Expression Profiling , Gene Silencing , Humans , Immunohistochemistry , Lipid Metabolism/genetics , Phospholipids/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Estrogen/metabolism , Survival Rate
18.
Scand J Pain ; 2(4): 155-160, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-29913754

ABSTRACT

Background and aims Burning mouth syndrome (BMS) and atypical facial pain (AFP) are often persistent idiopathic pain conditions that mainly affect middle-aged and elderly women. They have both been associated with various psychiatric disorders. This study examined current and lifetime prevalence of psychiatric axis I (symptom-based) and II (personality) disorders in patients with chronic idiopathic orofacial pain, and investigated the temporal relationship of psychiatric disorders and the onset of orofacial pain. Method Forty patients with BMS and 23 patients with AFP were recruited from Turku university hospital clinics. Mean age of the patients was 62.3 years (range 35-84) and 90% were female. BMS and AFP diagnoses were based on thorough clinical evaluation, and all patients had undergone clinical neurophysiological investigations including blink reflex and thermal quantitative tests. Current and lifetime DSM-IV diagnoses of axis I and II disorders were made on clinical basis with the aid of SCID-I and II-interviews. The detected prevalence rates and their 95% confidence intervals based on binomial distribution were compared to three previous large population-based studies. Results Of the 63 patients, 26 (41.3%) had had an axis I disorder that preceded the onset of orofacial pain, and 33 (52.4%) had had a lifetime axis I disorder. Rate of current axis I disorders was 36.5%, indicating that only about 16% of lifetime disorders had remitted, and they tended to run chronic course. The most common lifetime axis I disorders were major depression (30.2%), social phobia (15.9%), specific phobia (11.1%), and panic disorder (7.9%). Twelve patients (19.0%) had at least one cluster C personality disorder already before the emergence of orofacial pain. Patients with cluster C personality disorders are characterized as fearful and neurotic. None of the patients had cluster A (characterized as odd and eccentric) or B (characterized as dramatic, emotional or erratic) personality disorders. The most common personality disorders were obsessive-compulsive personality (14.3%), dependent personality (4.8%), and avoidant personality (3.2%). The majority of the patients (54%) had also one or more chronic pain conditions other than orofacial pain. In almost all patients (94%) they were already present at the onset of orofacial pain. Conclusions Our results suggest that major depression, persistent social phobia, and neurotic, fearful, and obsessive-compulsive personality characteristics are common in patients with chronic idiopathic orofacial pain. Most psychiatric disorders precede the onset of orofacial pain and they tend to run a chronic course. Implications We propose that the high psychiatric morbidity, and comorbidity to other chronic pain conditions, in chronic idiopathic orofacial pain can be best understood in terms of shared vulnerability to both chronic pain and specific psychiatric disorders, most likely mediated by dysfunctional brain dopamine activity.

19.
Neoplasia ; 12(11): 877-88, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21082043

ABSTRACT

Aneuploidy is among the most obvious differences between normal and cancer cells. However, mechanisms contributing to development and maintenance of aneuploid cell growth are diverse and incompletely understood. Functional genomics analyses have shown that aneuploidy in cancer cells is correlated with diffuse gene expression signatures and aneuploidy can arise by a variety of mechanisms, including cytokinesis failures, DNA endoreplication, and possibly through polyploid intermediate states. To identify molecular processes contributing to development of aneuploidy, we used a cell spot microarray technique to identify genes inducing polyploidy and/or allowing maintenance of polyploid cell growth in breast cancer cells. Of 5760 human genes screened, 177 were found to induce severe DNA content alterations on prolonged transient silencing. Association with response to DNA damage stimulus and DNA repair was found to be the most enriched cellular processes among the candidate genes. Functional validation analysis of these genes highlighted GINS2 as the highest ranking candidate inducing polyploidy, accumulation of endogenous DNA damage, and impairing cell proliferation on inhibition. The cell growth inhibition and induction of polyploidy by suppression of GINS2 was verified in a panel of breast cancer cell lines. Bioinformatic analysis of published gene expression and DNA copy number studies of clinical breast tumors suggested GINS2 to be associated with the aggressive characteristics of a subgroup of breast cancers in vivo. In addition, nuclear GINS2 protein levels distinguished actively proliferating cancer cells suggesting potential use of GINS2 staining as a biomarker of cell proliferation as well as a potential therapeutic target.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genomics/methods , Polyploidy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Cluster Analysis , Female , Humans , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
20.
Buenos Aires; Eudeba; 1a. ed; 1964. 311 p. 22 cm.(Temas de Eudeba Psicología). (77732).
Monography in Spanish | BINACIS | ID: bin-77732
SELECTION OF CITATIONS
SEARCH DETAIL
...