Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
J Pathol ; 258(3): 250-263, 2022 11.
Article in English | MEDLINE | ID: mdl-36148685

ABSTRACT

In melanoma, immunocytology (IC) after sentinel lymph node disaggregation not only enables better quantification of disseminated cancer cells (DCCs) than routine histopathology (HP) but also provides a unique opportunity to detect, isolate, and analyse these earliest harbingers of metachronous metastasis. Here, we explored lymph node IC in non-small cell lung cancer (NSCLC). For 122 NSCLC patients, 220 lymph nodes (LNs) were split in half and prepared for IC and HP. When both methods were compared, IC identified 22% positive patients as opposed to 4.5% by HP, revealing a much higher sensitivity of IC (p < 0.001). Assessment of all available 2,952 LNs of the same patients by HP uncovered additional patients escaping detection of lymphatic tumour spread by IC alone, consistent with the concept of skip metastasis. A combined lymph node status of IC and complete HP on a larger cohort of patients outperformed all risk factors in multivariable analysis for prognosis (p < 0.001; RR = 2.290; CI 1.407-3.728). Moreover, isolation of DCCs and single-cell molecular characterization revealed that (1) LN-DCCs differ from primary tumours in terms of copy number alterations and selected mutations and (2) critical alterations are acquired during colony formation within LNs. We conclude that LN-IC in NSCLC patients when combined with HP improves diagnostic precision, has the potential to reduce total workload, and facilitates molecular characterization of lymphatically spread cancer cells, which may become key for the selection and development of novel systemic therapies. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Evolution, Molecular , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Neoplasm Staging , Prognosis , Retrospective Studies
2.
Cancer Lett ; 499: 209-219, 2021 02 28.
Article in English | MEDLINE | ID: mdl-33276040

ABSTRACT

Diabetes mellitus type II (DM) and immune cell infiltration determine patient outcome in many tumor entities. Here we studied a possible link between the metabolic and immune cell status of OSCC patients. Glucose transporter (GLUT) 1 mRNA expression was elevated in all tumor samples, whereas other glycolytic markers such as lactate dehydrogenase (LDH) A or monocarboxylate transporter (MCT) 1 were increased in tumor samples from patients with diabetes and these patients had a significantly worse prognosis compared to non-diabetic patients. Analyses of immune cell infiltration in tumors from diabetic and non-diabetic patients revealed an increased leukocyte (CD45+) infiltration compared to normal mucosa only in non-diabetic patients. In line, the amount of CD3+ T cells per mm2 tumor tissue, was elevated in patients without diabetes and crucial for patient outcome in OSCC patients without diabetes, as compared to healthy mucosa using fluorescence immunohistochemistry in tissue microarrays of 229 patients. Our results demonstrate that diabetes is a prognostic factor for OSCC patients and associates with decreased leukocyte and CD3+ infiltration indicating that metabolic differences between diabetic and non-diabetic patients may alter tumor-infiltrating T cells and thereby determine patient outcome.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Lymphocytes, Tumor-Infiltrating/immunology , Mouth Neoplasms/mortality , Squamous Cell Carcinoma of Head and Neck/mortality , T-Lymphocytes/immunology , Adult , CD3 Complex/metabolism , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Glucose Transporter Type 1/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Monocarboxylic Acid Transporters/metabolism , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Mouth Mucosa/surgery , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/surgery , Predictive Value of Tests , Prognosis , Risk Assessment/methods , Risk Assessment/statistics & numerical data , Risk Factors , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/surgery , Symporters/metabolism , T-Lymphocytes/metabolism , Warburg Effect, Oncologic
3.
Nat Commun ; 11(1): 4977, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020483

ABSTRACT

Although thousands of breast cancer cells disseminate and home to bone marrow until primary surgery, usually less than a handful will succeed in establishing manifest metastases months to years later. To identify signals that support survival or outgrowth in patients, we profile rare bone marrow-derived disseminated cancer cells (DCCs) long before manifestation of metastasis and identify IL6/PI3K-signaling as candidate pathway for DCC activation. Surprisingly, and similar to mammary epithelial cells, DCCs lack membranous IL6 receptor expression and mechanistic dissection reveals IL6 trans-signaling to regulate a stem-like state of mammary epithelial cells via gp130. Responsiveness to IL6 trans-signals is found to be niche-dependent as bone marrow stromal and endosteal cells down-regulate gp130 in premalignant mammary epithelial cells as opposed to vascular niche cells. PIK3CA activation renders cells independent from IL6 trans-signaling. Consistent with a bottleneck function of microenvironmental DCC control, we find PIK3CA mutations highly associated with late-stage metastatic cells while being extremely rare in early DCCs. Our data suggest that the initial steps of metastasis formation are often not cancer cell-autonomous, but also depend on microenvironmental signals.


Subject(s)
Interleukin-6/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Bone Marrow/pathology , Breast/cytology , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Cytokine Receptor gp130/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Interleukin-6/genetics , Mutation , Neoplasm Metastasis/genetics , Receptors, Interleukin-6/deficiency , Receptors, Interleukin-6/metabolism , Stromal Cells/metabolism , Tumor Microenvironment
4.
Ann Surg ; 272(2): 366-376, 2020 08.
Article in English | MEDLINE | ID: mdl-32675551

ABSTRACT

OBJECTIVE: We aimed to define preoperative clinical and molecular characteristics that would allow better patient selection for operative resection. BACKGROUND: Although we use molecular selection methods for systemic targeted therapies, these principles are not applied to surgical oncology. Improving patient selection is of vital importance for the operative treatment of pancreatic cancer (pancreatic ductal adenocarcinoma). Although surgery is the only chance of long-term survival, 80% still succumb to the disease and approximately 30% die within 1 year, often sooner than those that have unresected local disease. METHOD: In 3 independent pancreatic ductal adenocarcinoma cohorts (total participants = 1184) the relationship between aberrant expression of prometastatic proteins S100A2 and S100A4 and survival was assessed. A preoperative nomogram based on clinical variables available before surgery and expression of these proteins was constructed and compared to traditional measures, and a postoperative nomogram. RESULTS: High expression of either S100A2 or S100A4 was independent poor prognostic factors in a training cohort of 518 participants. These results were validated in 2 independent patient cohorts (Glasgow, n = 198; Germany, n = 468). Aberrant biomarker expression stratified the cohorts into 3 distinct prognostic groups. A preoperative nomogram incorporating S100A2 and S100A4 expression predicted survival and nomograms derived using postoperative clinicopathological variables. CONCLUSIONS: Of those patients with a poor preoperative nomogram score, approximately 50% of patients died within a year of resection. Nomograms have the potential to improve selection for surgery and neoadjuvant therapy, avoiding surgery in aggressive disease, and justifying more extensive resections in biologically favorable disease.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Chemotactic Factors/genetics , Pancreatectomy/methods , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , S100 Proteins/genetics , Aged , Carcinoma, Pancreatic Ductal/surgery , Cause of Death , Cohort Studies , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Nomograms , Pancreatectomy/mortality , Pancreatic Neoplasms/surgery , Patient Selection , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Assessment , Survival Analysis
5.
Int J Mol Sci ; 21(5)2020 Mar 05.
Article in English | MEDLINE | ID: mdl-32150869

ABSTRACT

Patients with pancreatic ductal adenocarcinoma (PDAC) normally have a poor long-term prognosis. However, some rare cases of long-term survivors have been reported. The tumor microenvironment, consisting of cellular and stromal components, possibly plays an important role and might influence prognosis. In this context, the role of tumor-infiltrating B-cells and its impact on the survival in patients with PDAC remains controversial. We therefore aimed to assess the prognostic value of CD20-positive B-cells and CD20-positive B-cell aggregates as well as CD138, IgM, Pax5, and Ki67 on the survival of patients with PDAC using immunohistochemistry of FFPE pancreatectomy tissue sections from patients that underwent primary surgery for pT3- and R0-pancreatic adenocarcinoma between 1995 and 2016. Patients with PDAC were matched and grouped in 16 long-term-survivors (LTS, median overall survival (OS): 96 months [range: 61-177 months]) and 16 short-term-survivors (STS, median OS: 16 months [range: 7-32 months]). CD20-positive B-cells and B-cell aggregates in the tumor infiltration zone were significantly upregulated in the LTS-group compared to the STS-group (p = 0.0499 respectively p = 0.0432). Regarding the entire patient cohort (n = 32) CD20 positive B-cell aggregates in the tumor infiltration zone were an independent prognostic marker for overall survival in multivariate analysis (HR 9.2, CI 1.6-51.4, p = 0.012). These results underline the importance of tumor-associated B-cells for prognosis of patients with PDAC. The detailed role of B cells in the pathomechanism of PDAC should be further investigated for predicting outcome, identifying appropriate treatment regimens, and developing novel therapeutic options.


Subject(s)
Antigens, CD20/metabolism , B-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Pancreatectomy/mortality , Pancreatic Neoplasms/mortality , Tumor Microenvironment/immunology , Aged , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Prognosis , Survival Rate
6.
J Clin Apher ; 35(3): 163-171, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32163632

ABSTRACT

BACKGROUND: Homozygous familial hypercholesterolemia (hoFH) can cause severe atherosclerotic cardiovascular disease (ASCVD) in early infancy. Diagnosis and initiation of effective lipid-lowering therapy (LLT) are recommended as early as possible to prevent ASCVD-related morbidity and mortality. METHODS: The clinical courses of a pair of siblings with an identical hoFH genotype, who exhibited major similarities of their clinical phenotype were analyzed in a case-control fashion including the family. RESULTS: The older sibling was diagnosed with hoFH at the age of 4. Untreated LDL-cholesterol (LDL-C) was 17 mmol/L (660 mg/dL). LLT including lipoprotein apheresis (LA) was initiated and has been successful for 8 years now. A reduction of estimated cholesterol burden by 74% was achieved by LA and combined drug therapy including statins and ezetimibe. The efficacy of escalation of drug therapy was limited because the underlying LDL receptor (LDLR) mutation in the family resulted in substantially reduced receptor function. Treatment with proprotein convertase subtilisin-kexin type 9 (PCSK9)-antibodies failed. His younger brother died at the age of 2 years shortly after the hoFH diagnosis of the elder sibling. Postmortem examination revealed advanced aortic root atheroma and aortic valve stenosis. In the older sibling, aortic valve stenosis and insufficiency were treated at the age of 9 years with mechanical aortic valve replacement. CONCLUSIONS: LLT including LA should be initiated as early as possible following the diagnosis of hoFH with very high LDL-C levels. With the same genotype, the phenotype of hoFH can exhibit similar patterns but outcome is substantially related to treatment.


Subject(s)
Aortic Valve/physiopathology , Blood Component Removal/methods , Cholesterol, LDL/blood , Homozygote , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/therapy , Lipoproteins/therapeutic use , Adult , Aorta/pathology , Biopsy , Case-Control Studies , Child , Child, Preschool , Echocardiography , Family Health , Female , Genotype , Humans , Lipids/blood , Male , Phenotype , Proprotein Convertase 9/metabolism , Retrospective Studies , Siblings , Xanthomatosis/complications
7.
Front Oncol ; 9: 522, 2019.
Article in English | MEDLINE | ID: mdl-31275855

ABSTRACT

We have recently discovered that cancer cells take up extracellular citrate through plasma membrane citrate transporter (pmCiC) and advantageously use citrate for their metabolism. Citrate uptake can be blocked with gluconate and this results in decreased tumor growth and altered metabolic characteristics of tumor tissue. Interestingly, gluconate, considered to be physiologically neutral, is incidentally used in medicine as a cation carrier, but not as a therapeutically active substance. In this review we discuss the results of our recent research with available literature and suggest that gluconate may be useful in the treatment of cancer.

8.
EBioMedicine ; 40: 394-405, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30709769

ABSTRACT

BACKGROUND: Cytosolic 5'-nucleotidase 1A (NT5C1A) dephosphorylates non-cyclic nucleoside monophosphates to produce nucleosides and inorganic phosphates. Here, we investigate NT5C1A expression in pancreatic ductal adenocarcinoma (PDAC) and its impact on gemcitabine metabolism and therapeutic efficacy. METHODS: NT5C1A expression was determined by semiquantitative immunohistochemistry using tissue microarrays. Gemcitabine metabolites and response were assessed in several human and murine PDAC cell lines using crystal violet assays, Western blot, viability assays, and liquid chromatography tandem mass-spectrometry (LC-MS/MS). FINDINGS: NT5C1A was strongly expressed in tumor cells of a large subgroup of resected PDAC patients in two independent patient cohorts (44-56% score 2 and 8-26% score 3, n = 414). In contrast, NT5C1A was expressed at very low levels in the tumor stroma, and neither stromal nor tumoral expression was a prognostic marker for postoperative survival. In vitro, NT5C1A overexpression increased gemcitabine resistance by reducing apoptosis levels and significantly decreased intracellular amounts of cytotoxic dFdCTP in +NT5C1A tumor cells. Co-culture experiments with conditioned media from +NT5C1A PSCs improved gemcitabine efficacy in tumor cells. In vivo, therapeutic efficacy of gemcitabine was significantly decreased and serum levels of the inactive gemcitabine metabolite dFdU significantly increased in mice bearing NT5C1A overexpressing tumors. INTERPRETATION: NT5C1A is robustly expressed in tumor cells of resected PDAC patients. Moreover, NT5C1A mediates gemcitabine resistance by decreasing the amount of intracellular dFdCTP, leading to reduced tumor cell apoptosis and larger pancreatic tumors in mice. Further studies should clarify the role of NT5C1A as novel predictor for gemcitabine treatment response in patients with PDAC.


Subject(s)
5'-Nucleotidase/genetics , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Expression , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Models, Biological , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Prognosis , Xenograft Model Antitumor Assays , Gemcitabine
9.
Cancer Res ; 78(10): 2513-2523, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29510993

ABSTRACT

Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy.Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. Cancer Res; 78(10); 2513-23. ©2018 AACR.


Subject(s)
Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Cell Proliferation/drug effects , Citric Acid/metabolism , Gluconates/pharmacology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Pancreatic Neoplasms/pathology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Epithelial Cells/metabolism , Fatty Acids/biosynthesis , Glycolysis/physiology , Humans , Male , Mice , Organic Anion Transporters , Prostate/cytology , Prostate/metabolism , RNA Interference , RNA, Small Interfering/genetics
10.
Nat Commun ; 9(1): 595, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426936

ABSTRACT

Mouse models indicate that metastatic dissemination occurs extremely early; however, the timing in human cancers is unknown. We therefore determined the time point of metastatic seeding relative to tumour thickness and genomic alterations in melanoma. Here, we find that lymphatic dissemination occurs shortly after dermal invasion of the primary lesion at a median thickness of ~0.5 mm and that typical driver changes, including BRAF mutation and gained or lost regions comprising genes like MET or CDKNA2, are acquired within the lymph node at the time of colony formation. These changes define a colonisation signature that was linked to xenograft formation in immunodeficient mice and death from melanoma. Thus, melanoma cells leave primary tumours early and evolve at different sites in parallel. We propose a model of metastatic melanoma dormancy, evolution and colonisation that will inform direct monitoring of adjuvant therapy targets.


Subject(s)
Melanoma/genetics , Mutation , Skin Neoplasms/genetics , Skin/metabolism , Animals , Cell Line, Tumor , Comparative Genomic Hybridization/methods , Female , GTP Phosphohydrolases/genetics , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Melanoma/pathology , Membrane Proteins/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Proto-Oncogene Proteins p21(ras)/genetics , Skin/pathology , Skin Neoplasms/pathology , Transplantation, Heterologous
12.
PLoS One ; 12(1): e0170390, 2017.
Article in English | MEDLINE | ID: mdl-28114355

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDAC) is one of the deadliest tumors worldwide. Understanding the function of gene expression alterations is a prerequisite for developing new strategies in diagnostic and therapy. GPRC5A (RAI3), coding for a seven transmembrane G protein-coupled receptor is known to be overexpressed in pancreatic cancer and might be an interesting candidate for therapeutic intervention. Expression levels of RAI3 were compared using a tissue microarray of 435 resected patients with pancreatic cancer as well as 209 samples from chronic pancreatitis (CP), intra-ductal papillary mucinous neoplasm (IPMN) and normal pancreatic tissue. To elucidate the function of RAI3 overexpression, siRNA based knock-down was used and transfected cells were analyzed using proliferation and migration assays. Pancreatic cancer patients showed a statistically significant overexpression of RAI3 in comparison to normal and chronic pancreatitis tissue. Especially the loss of apical RAI3 expression represents an independent prognostic parameter for overall survival of patients with pancreatic cancer. Suppression of GPRC5a results in decreased cell growth, proliferation and migration in pancreatic cancer cell lines via a STAT3 modulated pathway, independent from ERK activation.


Subject(s)
Cell Proliferation , Pancreatic Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , STAT3 Transcription Factor/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Phosphorylation , Prognosis
13.
PLoS One ; 11(6): e0156900, 2016.
Article in English | MEDLINE | ID: mdl-27285579

ABSTRACT

OBJECTIVE: Despite substantial progress in recent years, graft survival beyond the first year still requires improvement. Since modern immunosuppression addresses mainly T-cell activation and proliferation, we studied macrophage infiltration into the allografts of 103 kidney transplant recipients during acute antibody and T-cell mediated rejection. Macrophage infiltration was correlated with both graft function and graft survival until month 36 after transplantation. RESULTS: Macrophage infiltration was significantly elevated in antibody-mediated and T-cell mediated rejection, but not in kidneys with established IFTA. Treatment of rejection with steroids was less successful in patients with more prominent macrophage infiltration into the allografts. Macrophage infiltration was accompanied by increased cell proliferation as well as antigen presentation. With regard to the compartmental distribution severity of T-cell-mediated rejection was correlated to the amount of CD68+ cells especially in the peritubular and perivascular compartment, whereas biopsies with ABMR showed mainly peritubular CD68 infiltration. Furthermore, severity of macrophage infiltration was a valid predictor of resulting creatinine values two weeks as well as two and three years after renal transplantation as illustrated by multivariate analysis. Additionally performed ROC curve analysis showed that magnitude of macrophage infiltration (below vs. above the median) was a valid predictor for the necessity to restart dialysis. Having additionally stratified biopsies in accordance to the magnitude of macrophage infiltration, differential CD68+ cell infiltration was reflected by striking differences in overall graft survival. CONCLUSION: The differences in acute allograft rejection have not only been reflected by different magnitudes of macrophage infiltration, but also by compartment-specific infiltration pattern and subsequent impact on resulting allograft function as well as need for dialysis initiation. There is a robust relationship between macrophage infiltration, accompanying antigen-presentation and resulting allograft function.


Subject(s)
Cell Movement , Graft Rejection/immunology , Graft vs Host Disease/immunology , Kidney Transplantation , Macrophages/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Allografts , Cell Movement/immunology , Female , Follow-Up Studies , Graft Rejection/diagnosis , Graft vs Host Disease/diagnosis , Humans , Male , Middle Aged , Prognosis , Severity of Illness Index , Transplantation Immunology , Treatment Outcome , Young Adult
14.
J Cancer ; 7(8): 915-20, 2016.
Article in English | MEDLINE | ID: mdl-27313781

ABSTRACT

Activating mutations in the core promoter of the TERT gene have been described in many different tumor entities. In vitro models showed a two- to fourfold increase in transcriptional activity of the TERT promoter through creation of a consensus binding motif for Ets/TCF transcription factors caused by these mutations. TERT core promoter mutations are the most common mutations in bladder cancer with a frequency between 55.6% and 82.8% described so far, and are independent of stage and grade. Since limited data on molecular alterations of early-onset bladder tumors exists, we assessed the frequency of TERT core promoter mutations in early-onset bladder cancer. Two cohorts of bladder tumors (early-onset patient group; n=144 (age of onset of disease ≤45 years); unselected, consecutive group; n=125) were examined for TERT core promoter mutations. After microdissection and extraction of DNA the corresponding hotspot regions in the TERT core promoter were examined by Sanger-sequencing or a SNaPshot approach. A significantly lower frequency of TERT core promoter mutations was found in tumors from the early-onset cohort compared to the consecutive cohort (57.6% vs. 84.8%, p<0.001). Among the early-onset cohort cases younger than the cohort's median age of 39 years at disease onset showed a significantly reduced number of TERT promoter mutations (31/67, 46,3%) than cases aged between 39 and 45 years (52/77, 67.5%; p=0.012). This association was not found in the consecutive cases. Mutation status was independent of tumor stage and grade. We conclude that in tumors from early-onset bladder cancer patients TERT core promoter mutations are not as frequent as in bladder tumors from consecutive cases, but seem to play an important role there as well. In patients below 39 years of age TERT core promoter mutations are a more infrequent event, suggesting different mechanisms of tumorigenesis in these young patients.

15.
Viszeralmedizin ; 31(3): 178-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26468312

ABSTRACT

BACKGROUND: ?Sclerosing cholangitis in critically ill patients' (SC-CIP) is a cholestatic liver disease of unknown etiology and represents the most prevalent form of secondary sclerosing cholangitis. METHODS: This overview is based on a systematic review of the literature searching for 'secondary sclerosing cholangitis', 'SC-CIP', 'cast syndrome', and 'ischemic cholangitis' in the database PubMed. RESULTS: SC-CIP can develop in patients with sepsis and acute respiratory distress syndrome during a long-term intensive care unit (ICU) treatment. It is a rare cholestatic liver disease with a rapid progression to liver cirrhosis and hepatic failure. SC-CIP is initiated by an ischemic injury to the biliary tree with subsequent stenoses of biliary ducts, biliary casts, and infections, often with multi-resistant bacteria. Mechanical ventilation with high positive end-expiratory pressure, prone positioning, and a higher volume of intraperitoneal fat have been proposed as risk factors for developing SC-CIP. Patients with SC-CIP have a poor prognosis, with liver transplantation (LT) being the only curative treatment option. CONCLUSION: In patients with sepsis, long-term ICU therapy and ongoing cholestasis SC-CIP must be excluded by endoscopic retrograde cholangiopancreatography. Due to the poor prognosis, the option of LT should be evaluated in all patients with SC-CIP.

16.
Inflamm Bowel Dis ; 21(9): 2039-51, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26083618

ABSTRACT

BACKGROUND: Longstanding ulcerative colitis (UC) bears a high risk for development of UC-associated colorectal carcinoma (UCC). The inflammatory microenvironment influences microRNA expression, which in turn deregulates target gene expression. microRNA-26b (miR-26b) was shown to be instrumental in normal tissue growth and differentiation. Thus, we aimed to investigate the impact of miR-26b in inflammation-associated colorectal carcinogenesis. METHODS: Two different cohorts of patients were investigated. In the retrospective group, a tissue microarray with 38 samples from 17 UC/UCC patients was used for miR-26b in situ hybridization and quantitative reverse transcription polymerase chain reaction analyses. In the prospective group, we investigated miR-26b expression in 25 fresh-frozen colon biopsies and corresponding serum samples of 6 UC and 15 non-UC patients, respectively. In silico analysis, Ago2-RNA immunoprecipitation, luciferase reporter assay, quantitative reverse transcription polymerase chain reaction examination, and miR-26b mimic overexpression were employed for target validation. RESULTS: miR-26b expression was shown to be upregulated with disease progression in tissues and serum of UC and UCC patients. Using miR-26b and Ki-67 expression levels, an UCC was predicted with high accuracy. We identified 4 novel miR-26b targets (DIP1, MDM2, CREBBP, BRCA1). Among them, the downregulation of the E3 ubiquitin ligase DIP1 was closely related to death-associated protein kinase stabilization along the normal mucosa-UC-UCC sequence. In silico functional pathway analysis revealed that the common cellular pathways affected by miR-26b are highly related to cancerogenesis and the development of gastrointestinal diseases. CONCLUSIONS: We suggest that miR-26b could serve as a biomarker for inflammation-associated processes in the gastrointestinal system. Because miR-26b expression is downregulated in sporadic colon cancer, it could discriminate between UCC and the sporadic cancer type.


Subject(s)
Biomarkers, Tumor/genetics , Carcinogenesis/metabolism , Colitis, Ulcerative/complications , Colorectal Neoplasms/genetics , MicroRNAs/metabolism , Adult , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , BRCA1 Protein/genetics , Biomarkers, Tumor/blood , CREB-Binding Protein/genetics , Celiac Disease/blood , Celiac Disease/complications , Child , Colitis, Ulcerative/blood , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/blood , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Ki-67 Antigen/metabolism , Luciferases , Male , Middle Aged , Prospective Studies , Proto-Oncogene Proteins c-mdm2/genetics , RNA/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
17.
Eur J Immunol ; 45(6): 1667-79, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25820779

ABSTRACT

Colorectal cancer (CRC) is one of the most common tumor entities. In patients with inflammatory bowel diseases, the development of colitis-associated colon cancer is considered a dangerous long-term complication. IL-17A and the transcription factor retinoic acid receptor-related orphan receptor γt (RORγt) play fundamental roles in the pathogenesis of inflammatory bowel diseases; in human studies, we detected a dense infiltration of RORγt-dependent CD4(+) IL17A(+) T helper (Th)17 cells in specimens of CRC, ulcerative colitis, and ulcerative colitis-associated colorectal cancer. However, the mechanistic role of RORγt(+) hematopoietic cells in colitis-associated tumorigenesis remains unclear. To investigate colitis-associated colon tumorigenesis, we conducted studies in the AOM+DSS mouse model that revealed the importance of RORγt for colon tumor progression. In the absence of RORγt-dependent Th17 lymphocytes, mice showed signs of intense chronic colitis, but developed significantly fewer macroscopic tumor nodules. The reduction of tumor development in RORγt(-/-) mice was not due to reduced colon tumor initiation. However, the proliferation rate of tumor cells was reduced in the absence of RORγt-dependent Th17 cells and tumor cells showed pronounced signs of senescence-associated epigenetic and lysosomal changes. These results indicate an important role for the immunological milieu in colitis-associated cancer, which is shaped in-part by RORγt-dependent Th17 lymphocytes that support CRC growth.


Subject(s)
Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Colitis/immunology , Colitis/metabolism , Colonic Neoplasms/etiology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Colitis/complications , Colitis/genetics , Colonic Neoplasms/pathology , Disease Models, Animal , Humans , Interleukin-17/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Tumor Burden/genetics , Tumor Burden/immunology
18.
Arch Toxicol ; 89(11): 2089-103, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25182422

ABSTRACT

In addition to its well-characterized role in the regulation of drug metabolism and transport by xenobiotics, pregnane X receptor (PXR) critically impacts on lipid homeostasis. In mice, both ligand-dependent activation and knockout of PXR were previously shown to promote hepatic steatosis. To elucidate the respective pathways in human liver, we generated clones of human hepatoma HepG2 cells exhibiting different PXR protein levels, and analyzed effects of PXR activation and knockdown on steatosis and expression of lipogenic genes. Ligand-dependent activation as well as knockdown of PXR resulted in increased steatosis in HepG2 cells. Activation of PXR induced the sterol regulatory element-binding protein (SREBP) 1-dependent lipogenic pathway via PXR-dependent induction of SREBP1a, which was confirmed in primary human hepatocytes. Inhibiting SREBP1 activity by blocking the cleavage-dependent maturation of SREBP1 protein impaired the induction of lipogenic SREBP1 target genes and triglyceride accumulation by PXR activation. On the other hand, PXR knockdown resulted in up-regulation of aldo-keto reductase (AKR) 1B10, which enhanced the acetyl-CoA carboxylase (ACC)-catalyzed reaction step of de novo lipogenesis. In a cohort of human liver samples histologically classified for non-alcoholic fatty liver disease, AKR1B10, SREBP1a and SREBP1 lipogenic target genes proved to be up-regulated in steatohepatitis, while PXR protein was reduced. In summary, our data suggest that activation and knockdown of PXR in human hepatic cells promote de novo lipogenesis and steatosis by induction of the SREBP1 pathway and AKR1B10-mediated increase of ACC activity, respectively, thus providing mechanistic explanations for a putative dual role of PXR in the pathogenesis of steatohepatitis.


Subject(s)
Fatty Liver/pathology , Hepatocytes/pathology , Receptors, Steroid/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Acetyl-CoA Carboxylase/metabolism , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Fatty Liver/genetics , Gene Knockdown Techniques , Gene Silencing , Hep G2 Cells , Humans , Ligands , Lipogenesis/genetics , Pregnane X Receptor , Receptors, Steroid/metabolism , Triglycerides/metabolism , Up-Regulation/genetics
19.
Nat Genet ; 47(1): 47-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485836

ABSTRACT

Here we describe a conditional piggyBac transposition system in mice and report the discovery of large sets of new cancer genes through a pancreatic insertional mutagenesis screen. We identify Foxp1 as an oncogenic transcription factor that drives pancreatic cancer invasion and spread in a mouse model and correlates with lymph node metastasis in human patients with pancreatic cancer. The propensity of piggyBac for open chromatin also enabled genome-wide screening for cancer-relevant noncoding DNA, which pinpointed a Cdkn2a cis-regulatory region. Histologically, we observed different tumor subentities and discovered associated genetic events, including Fign insertions in hepatoid pancreatic cancer. Our studies demonstrate the power of genetic screening to discover cancer drivers that are difficult to identify by other approaches to cancer genome analysis, such as downstream targets of commonly mutated human cancer genes. These piggyBac resources are universally applicable in any tissue context and provide unique experimental access to the genetic complexity of cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Transposable Elements/genetics , Gene Regulatory Networks , Mutagenesis, Insertional , Pancreatic Neoplasms/genetics , Amino Acid Sequence , Animals , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Knock-In Techniques , Genes, Synthetic , Genes, p16 , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Moths/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Proton-Translocating ATPases/genetics , RNA, Small Interfering/pharmacology , Repressor Proteins/analysis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Transgenes , Transposases/genetics , Transposases/physiology
20.
Cell Physiol Biochem ; 33(3): 692-704, 2014.
Article in English | MEDLINE | ID: mdl-24643085

ABSTRACT

BACKGROUND: Heart failure (CHF) is characterized by dyspnea and pulmonary changes. The underlying molecular adaptations are unclear, but might provide targets for therapeutic interventions. We therefore conceived a study to determine molecular changes of early pulmonary stress failure in a model of tachycardia-induced heart failure. METHODS: CHF was induced in rabbits by progessive right ventricular pacing (n=6). Invasive blood pressure measurements and echocardiography were repeatedly performed. Untreated animals served as controls (n=6). Pulmonary tissue specimens were subjected to two-dimensional gel electrophoresis, and differentially expressed proteins were identified by mass spectrometry. Selected proteins were validated by Western Blot analysis and localized by immunohistochemical staining. RESULTS: CHF animals were characterized by significantly altered functional, morphological, and hemodynamic parameters. Upon proteomic profiling, a total of 33 proteins was found to be differentially expressed in pulmonary tissue of CHF animals (18 up-regulated, and 15 down-regulated) belonging to 4 functional groups: 1. proteins involved in maintaining cytoarchitectural integrity, 2. plasma proteins indicating impaired alveolar-capillary permeability, 3. proteins with antioxidative properties, and 4. proteins participating in the metabolism of selenium compounds CONCLUSION: Experimental heart failure profoundly alters the pulmonary proteome. Our results supplement the current knowledge of pulmonary stress failure by specifying its molecular fundament.


Subject(s)
Heart Failure/metabolism , Lung/metabolism , Proteome/metabolism , Animals , Disease Models, Animal , Heart Failure/pathology , Lung/pathology , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...