Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Pathol ; 190(2): 323-332, 2020 02.
Article in English | MEDLINE | ID: mdl-31734230

ABSTRACT

Bacillus Calmette-Guérin instillation after removal of the tumor is the first line of treatment for urothelial carcinoma in situ (CIS), the precursor lesion of most muscle-invasive bladder cancers. Bacillus Calmette-Guérin therapy fails in >50% of cases, and second-line radical cystectomy is associated with overtreatment and drastic lifestyle consequences. Given the need for alternative bladder-preserving therapies, we identified genomic alterations (GAs) in urothelial CIS having the potential to predict response to targeted therapies. Laser-capture microdissection was applied to isolate 30 samples (25 CIS and 5 muscle controls) from 26 fresh-frozen cystectomy specimens. Targeted next-generation sequencing of 31 genes was performed. The panel comprised genes frequently affected in muscle-invasive bladder cancer of nonpapillary origin, focusing on potentially actionable GAs described to predict response to approved targeted therapies or drugs that are in registered clinical trials. Of CIS patients, 92% harbored at least one potentially actionable GA, which was identified in TP53/cell cycle pathway-related genes (eg, TP53 and MDM2) in 72%, genes encoding chromatin-modifying proteins (eg, ARID1A and KDM6A) in 68%, DNA damage repair genes (eg, BRCA2 and ATM) in 60%, and phosphatidylinositol 3-kinase/mitogen-activated protein kinase pathway genes (eg, ERBB2 and FGFR1) in 36% of the cases. These data might help guide the selection of targeted therapies to be investigated in future clinical CIS trials, and they may provide a basis for future mechanistic studies of urothelial CIS pathogenesis.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma in Situ/genetics , Cystectomy/methods , High-Throughput Nucleotide Sequencing/methods , Urinary Bladder Neoplasms/genetics , Urinary Bladder/metabolism , Aged , Aged, 80 and over , Carcinoma in Situ/pathology , Carcinoma in Situ/surgery , Case-Control Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery
2.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768978

ABSTRACT

Histologically, bladder cancer is a heterogeneous group comprising urothelial carcinoma (UC), squamous cell carcinoma, adenocarcinomas (ACs), urachal carcinomas (UrCs), and small cell neuroendocrine carcinomas (SCCs). However, all bladder cancers have been treated so far uniformly, and targeted therapy options are still limited. Thus, we aimed to determine the protein expression/molecular status of commonly used cancer targets (programmed cell death 1 ligand 1 (PD-L1), mismatch repair (MMR), androgen and estrogen receptors (AR/ER), Nectin-4, tumor-associated calcium signal transducer 2 (Tacstd2, Trop-2), epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and fibroblast growth factor receptor 3 (FGFR3)) to give first insights into whether patients with SCC, AC/UrCs, and squamous-differentiated carcinomas (Sq-BLCA) of the bladder could be eligible for targeted therapies. In addition, for MMR-deficient tumors, microsatellite instability was analyzed. We completed our own data with molecular data from The Cancer Genome Atlas (TCGA). We present ratios for each drug and cumulative ratios for multiple therapeutic options for each nonurothelial subtype. For example, 58.9% of SCC patients, 33.5% of AC/UrCs patients, and 79.3% of Sq-BLCA patients would be eligible for at least one of the analyzed targets. In conclusion, our findings hold promise for targeted therapeutic approaches in selected patients in the future, as various drugs could be applied according to the biomarker status.


Subject(s)
Molecular Targeted Therapy/methods , Urinary Bladder Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , DNA Mismatch Repair , Female , Humans , Immunohistochemistry , Male , Microsatellite Instability , Molecular Targeted Therapy/trends , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
3.
Pathologe ; 42(Suppl 1): 69-75, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33721057

ABSTRACT

BACKGROUND: Autopsy is an important tool for understanding the pathogenesis of diseases, including COVID-19. MATERIAL AND METHODS: On 15 April 2020, together with the German Society of Pathology and the Federal Association of German Pathologists, the German Registry of COVID-19 Autopsies (DeRegCOVID) was launched ( www.DeRegCOVID.ukaachen.de ). Building on this, the German Network for Autopsies in Pandemics (DEFEAT PANDEMIcs) was established on 1 September 2020. RESULTS: The main goal of DeRegCOVID is to collect and distribute de facto anonymized data on potentially all autopsies of people who have died from COVID-19 in Germany in order to meet the need for centralized, coordinated, and structured data collection and reporting during the pandemic. The success of the registry strongly depends on the willingness of the respective centers to report the data, which has developed very positively so far and requires special thanks to all participating centers. The rights to own data and biomaterials (stored decentrally) remain with each respective center. The DEFEAT PANDEMIcs network expands on this and aims to strengthen harmonization and standardization as well as nationwide implementation and cooperation in the field of pandemic autopsies. CONCLUSIONS: The extraordinary cooperation in the field of autopsies in Germany during the COVID-19 pandemic is impressively demonstrated by the establishment of DeRegCOVID, the merger of the registry of neuropathology (CNS-COVID19) with DeRegCOVID and the establishment of the autopsy network DEFEAT PANDEMIcs. It gives a strong signal for the necessity, readiness, and expertise to jointly help manage current and future pandemics by autopsy-derived knowledge.


Subject(s)
COVID-19 , Pandemics , Autopsy , Humans , Registries , SARS-CoV-2
4.
Pathologe ; 42(2): 216-223, 2021 Mar.
Article in German | MEDLINE | ID: mdl-33594614

ABSTRACT

BACKGROUND: Autopsy is an important tool for understanding the pathogenesis of diseases, including COVID-19. MATERIAL AND METHODS: On 15 April 2020, together with the German Society of Pathology and the Federal Association of German Pathologists, the German Registry of COVID-19 Autopsies (DeRegCOVID) was launched ( www.DeRegCOVID.ukaachen.de ). Building on this, the German Network for Autopsies in Pandemics (DEFEAT PANDEMIcs) was established on 1 September 2020. RESULTS: The main goal of DeRegCOVID is to collect and distribute de facto anonymized data on potentially all autopsies of people who have died from COVID-19 in Germany in order to meet the need for centralized, coordinated, and structured data collection and reporting during the pandemic. The success of the registry strongly depends on the willingness of the respective centers to report the data, which has developed very positively so far and requires special thanks to all participating centers. The rights to own data and biomaterials (stored decentrally) remain with each respective center. The DEFEAT PANDEMIcs network expands on this and aims to strengthen harmonization and standardization as well as nationwide implementation and cooperation in the field of pandemic autopsies. CONCLUSIONS: The extraordinary cooperation in the field of autopsies in Germany during the COVID-19 pandemic is impressively demonstrated by the establishment of DeRegCOVID, the merger of the registry of neuropathology (CNS-COVID19) with DeRegCOVID and the establishment of the autopsy network DEFEAT PANDEMIcs. It gives a strong signal for the necessity, readiness, and expertise to jointly help manage current and future pandemics by autopsy-derived knowledge.


Subject(s)
COVID-19 , Pandemics , Autopsy , Humans , Registries , SARS-CoV-2
5.
Pathologe ; 42(Suppl 1): 110-118, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34477921

ABSTRACT

Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD­1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI­H colorectal cancer (CRC). Further indications, such as dMMR/MSI­H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI­H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.


Subject(s)
Microsatellite Instability , Neoplasms/drug therapy , Neoplasms/genetics , DNA Mismatch Repair , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunohistochemistry , Prognosis
6.
Pathologe ; 42(4): 414-423, 2021 Jul.
Article in German | MEDLINE | ID: mdl-34043067

ABSTRACT

Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD­1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI­H colorectal cancer (CRC). Further indications, such as dMMR/MSI­H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI­H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.


Subject(s)
Colorectal Neoplasms , Microsatellite Instability , DNA Mismatch Repair , Humans , Immunohistochemistry , Prognosis
7.
BMC Cancer ; 20(1): 230, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32188412

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICI) are an integral part of bladder cancer therapy, however, the relevance of ICI treatment for mixed and pure squamous cell carcinoma of the bladder remains poorly studied. Therefore, we analysed the expression of programmed death-ligand 1 (PD-L1) in urothelial carcinomas with squamous differentiation (UC/SCC) and pure squamous cell carcinoma (SCC) of the bladder and studied a UC/SCC patient with ICI therapy. METHODS: Tissue microarrays of 45 UC/SCC and 63 SCC samples were immunohistochemically stained with four anti-PD-L1 antibodies (28-8, 22C3, SP142 and SP263). PD-L1 expression was determined for tumour cells (TP-Score), immune cells (IC-Score) and combined (CPS, combined positive score). In addition, we present clinical and histological data of an UC/SCC patient with nivolumab therapy. RESULTS: Overall, positive PD-L1 staining ranged between 4.8 and 61.9% for IC and 0 and 51.2% for TC depending on the used antibody. There were no significant differences between UC/SCC and SCC. According to current FDA guidelines for example for first line therapy of urothelial cancer with pembrolizumab (CPS ≥ 10), a subset of SCC patients up to 20% would be eligible. Finally, our UC/SCC index patient revealed excellent therapy response regarding his lung metastasis. CONCLUSIONS: Our data reveal a PD-L1 expression in squamous differentiated carcinomas comparable with current data shown for urothelial tumours. In accordance with the encouraging clinical data of the index patient we suggest ICI treatment also for mixed and pure SCC of the urinary bladder.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Squamous Cell/drug therapy , Nivolumab/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Tissue Array Analysis , Urinary Bladder Neoplasms/metabolism
8.
Int J Mol Sci ; 21(3)2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32046186

ABSTRACT

Bladder cancer is one of the more common malignancies in humans and the most expensive tumor for treating in the Unites States (US) and Europe due to the need for lifelong surveillance. Non-invasive tests approved by the FDA have not been widely adopted in routine diagnosis so far. Therefore, we aimed to characterize the two putative tumor suppressor genes ECRG4 and ITIH5 as novel urinary DNA methylation biomarkers that are suitable for non-invasive detection of bladder cancer. While assessing the analytical performance, a spiking experiment was performed by determining the limit of RT112 tumor cell detection (range: 100-10,000 cells) in the urine of healthy donors in dependency of the processing protocols of the RWTH cBMB. Clinically, urine sediments of 474 patients were analyzed by using quantitative methylation-specific PCR (qMSP) and Methylation Sensitive Restriction Enzyme (MSRE) qPCR techniques. Overall, ECRG4-ITIH5 showed a sensitivity of 64% to 70% with a specificity ranging between 80% and 92%, i.e., discriminating healthy, benign lesions, and/or inflammatory diseases from bladder tumors. When comparing single biomarkers, ECRG4 achieved a sensitivity of 73%, which was increased by combination with the known biomarker candidate NID2 up to 76% at a specificity of 97%. Hence, ITIH5 and, in particular, ECRG4 might be promising candidates for further optimizing current bladder cancer biomarker panels and platforms.


Subject(s)
Biomarkers, Tumor/urine , DNA Methylation , Proteinase Inhibitory Proteins, Secretory/genetics , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/urine , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/standards , Cell Line, Tumor , Female , Humans , Limit of Detection , Male , Middle Aged , Proteinase Inhibitory Proteins, Secretory/standards , Reproducibility of Results , Tumor Suppressor Proteins/standards , Urinary Bladder Neoplasms/diagnosis
9.
Int J Cancer ; 143(7): 1764-1773, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29672836

ABSTRACT

Urachal cancer (UrC) is a rare but aggressive malignancy often diagnosed in advanced stages requiring systemic treatment. Although cytotoxic chemotherapy is of limited effectiveness, prospective clinical studies can hardly be conducted. Targeted therapeutic treatment approaches and potentially immunotherapy based on a biological rationale may provide an alternative strategy. We therefore subjected 70 urachal adenocarcinomas to targeted next-generation sequencing, conducted in situ and immunohistochemical analyses (including PD-L1 and DNA mismatch repair proteins [MMR]) and evaluated the microsatellite instability (MSI) status. The analytical findings were correlated with clinicopathological and outcome data and Kaplan-Meier and univariable/multivariable Cox regression analyses were performed. The patients had a mean age of 50 years, 66% were male and a 5-year overall survival (OS) of 58% and recurrence-free survival (RFS) of 45% was detected. Sequence variations were observed in TP53 (66%), KRAS (21%), BRAF (4%), PIK3CA (4%), FGFR1 (1%), MET (1%), NRAS (1%), and PDGFRA (1%). Gene amplifications were found in EGFR (5%), ERBB2 (2%), and MET (2%). We detected no evidence of MMR-deficiency (MMR-d)/MSI-high (MSI-h), whereas 10 of 63 cases (16%) expressed PD-L1. Therefore, anti-PD-1/PD-L1 immunotherapy approaches might be tested in UrC. Importantly, we found aberrations in intracellular signal transduction pathways (RAS/RAF/PI3K) in 31% of UrCs with potential implications for anti-EGFR therapy. Less frequent potentially actionable genetic alterations were additionally detected in ERBB2 (HER2), MET, FGFR1, and PDGFRA. The molecular profile strengthens the notion that UrC is a distinct entity on the genomic level with closer resemblance to colorectal than to bladder cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Microsatellite Instability , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Adult , Aged , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Female , Follow-Up Studies , Gene Amplification , Gene Expression Profiling , Humans , Male , Middle Aged , Mutation , Prognosis , Young Adult
10.
Mol Carcinog ; 57(2): 167-181, 2018 02.
Article in English | MEDLINE | ID: mdl-28940371

ABSTRACT

ITIH5 has been proposed being a novel tumor suppressor in various tumor entities including breast cancer. Recently, ITIH5 was furthermore identified as metastasis suppressor gene in pancreatic carcinoma. In this study we aimed to specify the impact of ITIH5 on metastasis in breast cancer. Therefore, DNA methylation of ITIH5 promoter regions was assessed in breast cancer metastases using the TCGA portal and methylation-specific PCR (MSP). We reveal that the ITIH5 upstream promoter region is particularly responsible for ITIH5 gene inactivation predicting shorter survival of patients. Notably, methylation of this upstream ITIH5 promoter region was associated with disease progression, for example, abundantly found in distant metastases. In vitro, stably ITIH5-overexpressing MDA-MB-231 breast cancer clones were used to analyze cell invasion and to identify novel ITIH5-downstream targets. Indeed, ITIH5 re-expression suppresses invasive growth of MDA-MB-231 breast cancer cells while modulating expression of genes involved in metastasis including Endoglin (ENG), an accessory TGF-ß receptor, which was furthermore co-expressed with ITIH5 in primary breast tumors. By performing in vitro stimulation of TGF-ß signaling using TGF-ß1 and BMP-2 we show that ITIH5 triggered a TGF-ß superfamily signaling switch contributing to downregulation of targets like Id1, known to endorse metastasis. Moreover, ITIH5 predicts longer overall survival (OS) only in those breast tumors that feature high ENG expression or inversely regulated ID1 suggesting a clinical and functional impact of an ITIH5-ENG axis for breast cancer progression. Hence, we provide evidence that ITIH5 may represent a novel modulator of TGF-ß superfamily signaling involved in suppressing breast cancer metastasis.


Subject(s)
Breast Neoplasms/genetics , Endoglin/genetics , Proteinase Inhibitory Proteins, Secretory/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Methylation/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Genes, Tumor Suppressor/physiology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic/genetics , Risk
11.
Mol Cancer ; 16(1): 44, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28231808

ABSTRACT

BACKGROUND: Extracellular matrix (ECM) is known to maintain epithelial integrity. In carcinogenesis ECM degradation triggers metastasis by controlling migration and differentiation including cancer stem cell (CSC) characteristics. The ECM-modulator inter- α-trypsin inhibitor heavy chain family member five (ITIH5) was recently identified as tumor suppressor potentially involved in impairing breast cancer progression but molecular mechanisms underlying its function are still elusive. METHODS: ITIH5 expression was analyzed using the public TCGA portal. ITIH5-overexpressing single-cell clones were established based on T47D and MDA-MB-231 cell lines. Colony formation, growth, apoptosis, migration, matrix adhesion, traction force analyses and polarization of tumor cells were studied in vitro. Tumor-initiating characteristics were analyzed by generating a metastasis mouse model. To identify ITIH5-affected pathways we utilized genome wide gene expression and DNA methylation profiles. RNA-interference targeting the ITIH5-downstream regulated gene DAPK1 was used to confirm functional involvement. RESULTS: ITIH5 loss was pronounced in breast cancer subtypes with unfavorable prognosis like basal-type tumors. Functionally, cell and colony formation was impaired after ITIH5 re-expression in both cell lines. In a metastasis mouse model, ITIH5 expressing MDA-MB-231 cells almost completely failed to initiate lung metastases. In these metastatic cells ITIH5 modulated cell-matrix adhesion dynamics and altered biomechanical cues. The profile of integrin receptors was shifted towards ß1-integrin accompanied by decreased Rac1 and increased RhoA activity in ITIH5-expressing clones while cell polarization and single-cell migration was impaired. Instead ITIH5 expression triggered the formation of epithelial-like cell clusters that underwent an epigenetic reprogramming. 214 promoter regions potentially marked with either H3K4 and /or H3K27 methylation showed a hyper- or hypomethylated DNA configuration due to ITIH5 expression finally leading to re-expression of the tumor suppressor DAPK1. In turn, RNAi-mediated knockdown of DAPK1 in ITIH5-expressing MDA-MB-231 single-cell clones clearly restored cell motility. CONCLUSIONS: Our results provide evidence that ITIH5 triggers a reprogramming of breast cancer cells with known stem CSC properties towards an epithelial-like phenotype through global epigenetic changes effecting known tumor suppressor genes like DAPK1. Therewith, ITIH5 may represent an ECM modulator in epithelial breast tissue mediating suppression of tumor initiating cancer cell characteristics which are thought being responsible for the metastasis of breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Death-Associated Protein Kinases/genetics , Lung Neoplasms/secondary , Proteinase Inhibitory Proteins, Secretory/genetics , Animals , Cell Line, Tumor , Epigenesis, Genetic , Extracellular Matrix , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice , Neoplasm Transplantation , Prognosis , Survival Analysis
12.
Angiogenesis ; 19(4): 525-35, 2016 10.
Article in English | MEDLINE | ID: mdl-27582011

ABSTRACT

Renal microvascular rarefaction characterizes chronic kidney disease (CKD). In murine models of CKD, micro-CT imaging reflected capillary rarefaction using quantification of renal relative blood volume (rBV). In addition, micro-CT imaging revealed morphological alterations of the intrarenal vasculature including reduced vascular branching and lumen diameter. Here, we retrospectively quantified rBV in contrast-enhanced CT angiography in patients and found that, compared to non-CKD patients, those with CKD and renal fibrosis had significantly reduced rBV in the renal cortex. rBV values closely mirrored capillary rarefaction in the corresponding nephrectomy specimens. In patients with follow-up CT angiography, reduction of renal function was paralleled by a decline in rBV. Using virtual autopsy, i.e., postmortem CT angiography, morphometry of intrarenal arteries in 3D-rendered CT images revealed significantly reduced arterial diameter and branching in CKD compared to non-CKD cases. In conclusion, in CKD patients, contrast-enhanced CT imaging with quantification of rBV correlates with functional renal vasculature, whereas virtual autopsy allows morphometric analyses of macrovascular changes. Importantly, the observed vascular alterations in CKD patients mirror those in animals with progressive CKD, suggesting a high relevance of animal models for studying vascular alterations in CKD and renal fibrosis.


Subject(s)
Computed Tomography Angiography/methods , Renal Insufficiency, Chronic/diagnostic imaging , Aged , Animals , Blood Volume , Capillaries/diagnostic imaging , Capillaries/pathology , Cohort Studies , Contrast Media , Disease Progression , Fibrosis , Humans , Imaging, Three-Dimensional , Kidney/blood supply , Kidney/diagnostic imaging , Kidney/pathology , Male , Microvessels/diagnostic imaging , Microvessels/pathology , Middle Aged , Renal Circulation , Renal Insufficiency, Chronic/pathology , Retrospective Studies
13.
Strahlenther Onkol ; 192(7): 440-8, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27272660

ABSTRACT

BACKGROUND: Despite improved biochemical recurrence-free survival rates by the use of immediate adjuvant radiotherapy (RT) in patients with locally advanced prostate cancer, disagreement about the need and timing of RT remains. PATIENTS AND METHODS: From 2005-2009, 94 patients presenting with a stage pT3a N0 and microscopic positive resection margin were retrospectively analyzed after radical prostatectomy. Special attention was given to patients' outcome, the frequency of additive RT, and its efficacy. RESULTS: Median follow-up was 80 months. A total of 71 patients had a negative postoperative prostate-specific antigen (PSA) level (<0.07 ng/ml). Thirty-six of them did not experience any PSA relapse (subgroup 1). Fourteen of them received additive RT and during follow-up all 36 patients remained PSA negative. Of 71 initially PSA-negative patients, 35 had a biochemical relapse (subgroup 2); 28 patients underwent salvage RT. The median PSA value before salvage RT was 0.24 ng/ml and was subsequently negative (<0.07 ng/ml) in 23 patients after RT. Of the entire cohort, 23 patients had persisting PSA after surgery (subgroup 3). Of these, 18 patients received salvage RT at a median PSA level of 0.4 ng/ml. One patient in subgroup 1, 5 patients in subgroup 2, and 9 patients in subgroup 3 had ongoing androgen deprivation therapy. CONCLUSION: The present study of 94 pT3a N0 R1 prostate cancer patients provides insight into medical care of this patient cohort and underlines the need for additive RT for the majority of patients to achieve long-term biochemical control. Although immediate adjuvant RT was applied with restraint (20 %), during the observation period 60 of 94 patients (63.8 %) received RT - highlighting the need of postoperative treatment.


Subject(s)
Margins of Excision , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/prevention & control , Prostatic Neoplasms/blood , Prostatic Neoplasms/therapy , Radiotherapy, Conformal/methods , Aged , Biomarkers, Tumor/blood , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/pathology , Radiotherapy, Adjuvant/methods , Time-to-Treatment , Treatment Outcome
14.
Exp Dermatol ; 24(9): 663-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25809190

ABSTRACT

Inter-α-trypsin inhibitors are protease inhibitors that are thought to be important regulators in various acute-phase processes. They are composed of one light chain (bikunin) and different heavy chains (ITIHs). The only function known so far of ITIHs is the covalent linkage to hyaluronan (HA). As there is virtually no knowledge on the distribution and function of ITIH proteins in skin tissue, we performed a systematic characterization of ITIH expression in healthy and diseased skin. Using GeneChip(®) Human Exon 1.0 ST expression profiling, we found that ITIH5 represents the major ITIH family member expressed in human skin. Moreover, the use of quantitative reverse transcription PCR and a customized ITIH5-specific antibody indicated that ITIH5 is predominantly produced by dermal fibroblasts. Immunohistochemical analysis revealed a clearly detectable ITIH5 protein expression in normal skin. Interestingly, ITIH5 expression was significantly up-regulated in inflammatory skin diseases. Furthermore, 3D skin models employing murine Itih5(-/-) epidermal keratinocytes and dermal fibroblasts as well as skin specimens of Itih5(-/-) mice revealed a significantly altered epidermal structure compared to wild-type controls. Hence, we can strengthen the presumption that ITIH5 may constitute a novel regulatory molecule of the human skin that could play an important role in inflammation via its interaction with HA.


Subject(s)
Epidermis/metabolism , Gene Expression , Proteinase Inhibitory Proteins, Secretory/genetics , Proteinase Inhibitory Proteins, Secretory/metabolism , Skin Diseases/genetics , Skin Diseases/metabolism , Alpha-Globulins/genetics , Animals , Blood Proteins/genetics , Cells, Cultured , Epidermis/chemistry , Epidermis/pathology , Female , Fibroblasts , Gene Expression Profiling , Glycoproteins/genetics , Humans , Hyaluronic Acid/metabolism , Inflammation/genetics , Keratinocytes , Mice , Mice, Knockout , Models, Anatomic , Oligonucleotide Array Sequence Analysis , Proteinase Inhibitory Proteins, Secretory/analysis , RNA, Messenger/metabolism , Up-Regulation
15.
Carcinogenesis ; 35(3): 727-36, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24265292

ABSTRACT

Inter-α-trypsin inhibitor heavy chain 5 (ITIH5) has been associated with tumour suppression in various cancers. However, its putative role in bladder cancer is completely unknown. Therefore, we initiated a study analysing ITIH5 expression as well as its prognostic and functional impact on human urothelial cancers (UCs). Expression analysis showed a clear down-regulation of ITIH5 mRNA in 61% (n = 45) of UCs, especially in muscle-invasive tumours (P < 0.001). ITIH5 loss in UCs was further evident on protein level (65.5%, n = 55) as detected by immunohistochemistry. DNA methylation analysis demonstrated tumour-specific ITIH5 promoter methylation in 50% of papillary none-invasive pTa (n = 30) and 68% of invasive (n = 28) UCs. Aberrant ITIH5 promoter methylation in bladder tumours was tightly linked (P < 0.001) with loss of ITIH5 mRNA expression, which was furthermore functionally confirmed by demethylation analysis in cell lines. Pyrosequencing analysis revealed that ITIH5 promoter hypermethylation was closely associated with progressive bladder cancers. Subsequently, a large cohort (n = 120) of clinically challenging pT1 high-grade UC was analysed for ITIH5 expression. Of clinical significance, we found an association between loss of ITIH5 expression and unfavourable prognosis of UC patients without distant metastasis at first diagnosis (recurrence-free survival; hazard ratio: 4.35, P = 0.048). Functionally, ITIH5 re-expression in human RT112 bladder cancer cells led to both suppression of cell migration and inhibition of colony spreading. Hence, we provide evidence that down-regulation of ITIH5 by aberrant DNA hypermethylation may provoke invasive phenotypes in human bladder cancer. Moreover, ITIH5 protein might become a prognostic biomarker for relapse risk stratification in high-grade UC patients.


Subject(s)
Biomarkers, Tumor/genetics , Epigenesis, Genetic , Gene Silencing , Neoplasm Recurrence, Local/genetics , Proteinase Inhibitory Proteins, Secretory/genetics , Urinary Bladder Neoplasms/genetics , DNA Methylation , Disease Progression , Humans , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Urinary Bladder Neoplasms/pathology
16.
BMC Cancer ; 14: 901, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25465919

ABSTRACT

BACKGROUND: Posttranslational protein modifications are known to modulate key biological processes like proliferation and apoptosis. Accumulating evidence shows that ST6GAL1, an enzyme that catalyzes the transfer of sialic acid onto galactose-containing substrates, is aberrantly expressed in various cancers and may affect cell motility and invasion. This is the first study to describe ST6GAL1 expression and regulation in human bladder cancer. METHODS: ST6GAL1 mRNA expression levels in human cell lines (UROtsa, RT4, RT112 and J82) and tissue samples (n = 15 normal urothelium (NU), n = 13 papillary non-invasive tumors (pTa), n = 12 carcinoma in situ (CIS), n = 26 muscle invasive tumors (pT2-4)) were assessed using real-time PCR. In addition, ST6GAL1 protein expression was evaluated using immunohistochemistry. Promoter methylation analysis was performed using methylation-specific PCR (MSP) in cell lines (n = 4) and patient samples (n = 23 NU, n = 12 CIS, n = 29 pTa, n = 41 pT2-4). Epigenetic ST6GAL1 gene silencing was confirmed by in vitro demethylation of bladder cell lines. Data were validated by analysis of an independent bladder tumor data set (n = 184) based on The Cancer Genome Atlas (TCGA) portal. RESULTS: Semi-quantitative ST6GAL1 real-time PCR expression analysis showed two distinct trends: In muscle-invasive tumors ST6GAL1 expression was downregulation by 2.7-fold, while papillary non-invasive tumors showed an increased ST6GAL1 mRNA expression compared to normal urothelium. ST6GAL1 loss in muscle-invasive tumors was associated with increasing invasiveness. On the protein level, 69.2% (n = 45/65) of all tumors showed a weak ST6GAL1 protein staining (IRS ≤ 4) while 25.6% (16/65) exhibited a complete loss (IRS = 0) of ST6GAL1 protein. Tumor-specific DNA methylation of the ST6GAL1 promoter region was frequently found in pT2-4 tumors (53.6% (22/41)), whereas only 13.8% (4/29) of pTa tumors showed ST6GAL1 promoter methylation. Normal urothelium remained unmethylated. Importantly, we significantly revealed an inverse correlation between ST6GAL1 mRNA expression and ST6GAL1 promoter merthylation in primary bladder cancer. These findings were clearly verified by the TCGA public data set and in vitro demethylation assays functionally confirmed ST6GAL1 promoter methylation as a potential regulatory factor for ST6GAL1 gene silencing. CONCLUSIONS: Our study characterizes for the first time ST6GAL1 expression loss caused by aberrant ST6GAL1 promoter methylation potentially indicating a tumor suppressive role in bladder carcinogenesis.


Subject(s)
Antigens, CD/genetics , Carcinoma in Situ/genetics , Carcinoma, Papillary/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Sialyltransferases/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Line, Tumor , DNA Methylation , Female , Humans , Male , Middle Aged , Promoter Regions, Genetic , Sialyltransferases/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
17.
Mol Oncol ; 18(6): 1397-1416, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38429970

ABSTRACT

The effect of grainyhead-like transcription factor 3 (GRHL3) on cancer development depends on the cancer subtypes as shown in tumor entities such as colorectal or oral squamous cell carcinomas. Here, we analyzed the subtype-specific role of GRHL3 in bladder carcinogenesis, comparing common urothelial carcinoma (UC) with squamous bladder cancer (sq-BLCA). We examined GRHL3 mRNA and protein expression in cohorts of patient samples, its prognostic role and its functional impact on tumorigeneses in different molecular and histopathological subtypes of bladder cancer. We showed for GRHL3 a reverse expression in squamous and urothelial bladder cancer subtypes. Stably GRHL3-overexpressing EJ28, J82, and SCaBER in vitro models revealed a tumor-suppressive function in squamous and an oncogenic role in the urothelial cancer cells affecting cell and colony growth, and migratory and invasive capacities. Transcriptomic profiling demonstrated highly subtype-specific GRHL3-regulated expression networks coined by the enrichment of genes involved in integrin-mediated pathways. In SCaBER, loss of ras homolog family member A (RHOA) GTPase activity was demonstrated to be associated with co-regulation of eukaryotic translation initiation factor 4E family member 3 (EIF4E3), a potential tumor suppressor gene. Thus, our data provide for the first time a detailed insight into the role of the transcription factor GRHL3 in different histopathological subtypes of bladder cancer.


Subject(s)
Carcinogenesis , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Transcription Factors , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Line, Tumor , Carcinogenesis/genetics , Carcinogenesis/pathology , Female , Male , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Prognosis , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Aged
18.
Breast Cancer Res ; 15(1): R4, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23320751

ABSTRACT

INTRODUCTION: For early detection of breast cancer, the development of robust blood-based biomarkers that accurately reflect the host tumor is mandatory. We investigated DNA methylation in circulating free DNA (cfDNA) from blood of breast cancer patients and matched controls to establish a biomarker panel potentially useful for early detection of breast cancer. METHODS: We examined promoter methylation of seven putative tumor-suppressor genes (SFRP1, SFRP2, SFRP5, ITIH5, WIF1, DKK3, and RASSF1A) in cfDNA extracted from serum. Clinical performance was first determined in a test set (n = 261 sera). In an independent validation set (n = 343 sera), we validated the most promising genes for further use in early breast cancer detection. Sera from 59 benign breast disease and 58 colon cancer patients were included for additional specificity testing. RESULTS: Based on the test set, we determined ITIH5 and DKK3 promoter methylation as candidate biomarkers with the best sensitivity and specificity. In both the test and validation set combined, ITIH5 and DKK3 methylation achieved 41% sensitivity with a specificity of 93% and 100% in healthy and benign disease controls, respectively. Combination of these genes with RASSF1A methylation increased the sensitivity to 67% with a specificity of 69% and 82% in healthy controls and benign disease controls, respectively. CONCLUSIONS: Tumor-specific methylation of the three-gene panel (ITIH5, DKK3, and RASSF1A) might be a valuable biomarker for the early detection of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Intercellular Signaling Peptides and Proteins/blood , Proteinase Inhibitory Proteins, Secretory/blood , Tumor Suppressor Proteins/blood , Adaptor Proteins, Signal Transducing , Adult , Aged , Biomarkers, Tumor/blood , Breast Neoplasms/pathology , Chemokines , DNA Methylation/genetics , Early Detection of Cancer , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Promoter Regions, Genetic
19.
Nephrol Dial Transplant ; 28(4): 856-68, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23223222

ABSTRACT

BACKGROUND: Calcific uraemic arteriolopathy (CUA) or calciphylaxis is a rare, life-threatening disease predominantly occurring in patients with end-stage renal disease. Its pathogenesis has been suggested to include ectopic osteogenesis in soft tissue and the vasculature associated with extracellular matrix (ECM) remodelling. METHODS: To gain further insights into the pathogenesis of CUA, we performed systematic analyses of skin specimens obtained from seven CUA patients including histology, immunohistochemistry, electron microscopy, electron dispersive X-ray analysis (EDX) and quantitative real-time RT-PCR. Skin specimens of (i) seven patients without chronic kidney disease and without CUA and (ii) seven dialysis patients without CUA served as controls. RESULTS: In the CUA skin lesions, we observed a significant upregulation of bone morphogenic protein 2 (BMP-2), its target gene Runx2 and its indirect antagonist sclerostin. Furthermore, we detected an increased expression of inactive uncarboxylated matrix Gla protein (Glu-MGP). The upregulation of osteogenesis-associated markers was accompanied by an increased expression of osteopontin, fibronectin, laminin and collagen I indicating an extensive remodelling of the subcutaneous ECM. EDX analysis revealed calcium/phosphate accumulations in the subcutis of all CUA patients with a molar ratio of 1.68 ± 0.06 matching that of hydroxyapatite mineral. Widespread media calcification in cutaneous arterioles was associated with destruction of the endothelial layer and partial exfoliation of the endothelial cells (ECs). CD31 immunostaining revealed aggregates of ECs contributing to intraluminal obstruction and consecutive malperfusion resulting in the clinical picture of ulcerative necrosis in all seven patients. CONCLUSIONS: Our data indicate that CUA is an active osteogenic process including the upregulation of BMP-2 signalling, hydroxyapatite deposition and extensive matrix remodelling of the subcutis.


Subject(s)
Calciphylaxis/pathology , Calcium/metabolism , Extracellular Matrix/metabolism , Kidney Failure, Chronic/pathology , Osteogenesis/physiology , Skin Diseases/pathology , Uremia/pathology , Adult , Aged , Apoptosis , Blotting, Western , Calciphylaxis/metabolism , Case-Control Studies , Cell Proliferation , Cell Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoenzyme Techniques , Kidney Failure, Chronic/metabolism , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin Diseases/metabolism , Uremia/metabolism , Young Adult
20.
Nephrol Dial Transplant ; 28(4): 889-900, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23229927

ABSTRACT

BACKGROUND: Platelet-derived growth factors (PDGF)-AA and -CC mediate renal fibroblast proliferation and/or renal fibrosis. Whereas PDGF-CC binds to both the PDGF receptors (PDGFRs)-αα- and -αß, PDGF-AA binds more selectively to the αα-receptor, suggesting potential differences in the biological activities. METHODS: We compared signal transduction, gene expression as well as changes in the proteome induced by PDGF-AA and -CC in rat renal fibroblasts, which express both PDGFR subunits. The growth factor concentrations used were chosen based on their equipotency in inducing rat renal fibroblast proliferation. RESULTS: Both PDGF-AA and PDGF-CC induced phosphorylation and activation of extracellular signal-regulated kinase 1 (ERK1) and ERK2. Renal fibroblast proliferation induced by either PDGF-AA or -CC could be blocked by signal transduction inhibitors of the mitogen-activated protein kinase (MAPK)-, Janus-kinase (JAK)/signal transducers and activators of transcription (STAT) and phosphatidyl-inositol-3-kinase (PI3K) pathway, pointing to the involvement of all the three pathways. However, quantitative differences between both the stimulations were minor. Additive or synergistic effects by stimulating simultaneously with PDGF-AA and -CC were not observed. Using a proteomic approach we found eleven differentially expressed proteins, which were quantitatively altered after treatment with either PDGF-AA or PDGF-CC. The regulation of calreticulin and inorganic pyrophosphatase 1 could be verified by western blotting. CONCLUSIONS: PDGF-AA and -CC exhibit almost identical biological effects on signal transduction and proteome in cultured renal fibroblasts, suggesting that the ligands exert their activity essentially through the commonly bound PDGFR-αα. Nonetheless, two differentially expressed proteins were identified which might be involved in the development of renal failure.


Subject(s)
Fibroblasts/metabolism , Kidney/metabolism , Lymphokines/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Electrophoretic Mobility Shift Assay , Fibroblasts/cytology , Kidney/cytology , Lymphokines/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation , Platelet-Derived Growth Factor/genetics , Proteomics , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL