Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 158(1): 25-40, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995976

ABSTRACT

Obesity and diabetes affect more than half a billion individuals worldwide. Interestingly, the two conditions do not always coincide and the molecular determinants of "healthy" versus "unhealthy" obesity remain ill-defined. Chronic metabolic inflammation (metaflammation) is believed to be pivotal. Here, we tested a hypothesized anti-inflammatory role for heme oxygenase-1 (HO-1) in the development of metabolic disease. Surprisingly, in matched biopsies from "healthy" versus insulin-resistant obese subjects we find HO-1 to be among the strongest positive predictors of metabolic disease in humans. We find that hepatocyte and macrophage conditional HO-1 deletion in mice evokes resistance to diet-induced insulin resistance and inflammation, dramatically reducing secondary disease such as steatosis and liver toxicity. Intriguingly, cellular assays show that HO-1 defines prestimulation thresholds for inflammatory skewing and NF-κB amplification in macrophages and for insulin signaling in hepatocytes. These findings identify HO-1 inhibition as a potential therapeutic strategy for metabolic disease.


Subject(s)
Heme Oxygenase-1/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Obesity/complications , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Liver/metabolism , Macrophages/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Mice , Mice, Knockout , Obesity/physiopathology , Reactive Oxygen Species/metabolism
2.
Mol Cell ; 81(18): 3848-3865.e19, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547241

ABSTRACT

Metabolic rewiring and redox balance play pivotal roles in cancer. Cellular senescence is a barrier for tumorigenesis circumvented in cancer cells by poorly understood mechanisms. We report a multi-enzymatic complex that reprograms NAD metabolism by transferring reducing equivalents from NADH to NADP+. This hydride transfer complex (HTC) is assembled by malate dehydrogenase 1, malic enzyme 1, and cytosolic pyruvate carboxylase. HTC is found in phase-separated bodies in the cytosol of cancer or hypoxic cells and can be assembled in vitro with recombinant proteins. HTC is repressed in senescent cells but induced by p53 inactivation. HTC enzymes are highly expressed in mouse and human prostate cancer models, and their inactivation triggers senescence. Exogenous expression of HTC is sufficient to bypass senescence, rescue cells from complex I inhibitors, and cooperate with oncogenic RAS to transform primary cells. Altogether, we provide evidence for a new multi-enzymatic complex that reprograms metabolism and overcomes cellular senescence.


Subject(s)
Cellular Senescence/physiology , NAD/metabolism , Aging/metabolism , Aging/physiology , Animals , Cell Line, Tumor , Cellular Senescence/genetics , Cytosol , Glucose/metabolism , Humans , Hydrogen/chemistry , Hydrogen/metabolism , Malate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , NAD/physiology , Oxidation-Reduction , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism
3.
Nature ; 589(7842): 442-447, 2021 01.
Article in English | MEDLINE | ID: mdl-33361811

ABSTRACT

Successful pregnancies rely on adaptations within the mother1, including marked changes within the immune system2. It has long been known that the thymus, the central lymphoid organ, changes markedly during pregnancy3. However, the molecular basis and importance of this process remain largely obscure. Here we show that the osteoclast differentiation receptor RANK4,5 couples female sex hormones to the rewiring of the thymus during pregnancy. Genetic deletion of Rank (also known as Tnfrsf11a) in thymic epithelial cells results in impaired thymic involution and blunted expansion of natural regulatory T (Treg) cells in pregnant female mice. Sex hormones, in particular progesterone, drive the development of thymic Treg cells through RANK in a manner that depends on AIRE+ medullary thymic epithelial cells. The depletion of Rank in the mouse thymic epithelium results in reduced accumulation of natural Treg cells in the placenta, and an increase in the number of miscarriages. Thymic deletion of Rank also results in impaired accumulation of Treg cells in visceral adipose tissue, and is associated with enlarged adipocyte size, tissue inflammation, enhanced maternal glucose intolerance, fetal macrosomia, and a long-lasting transgenerational alteration in glucose homeostasis, which are all key hallmarks of gestational diabetes. Transplantation of Treg cells rescued fetal loss, maternal glucose intolerance and fetal macrosomia. In human pregnancies, we found that gestational diabetes also correlates with a reduced number of Treg cells in the placenta. Our findings show that RANK promotes the hormone-mediated development of thymic Treg cells during pregnancy, and expand the functional role of maternal Treg cells to the development of gestational diabetes and the transgenerational metabolic rewiring of glucose homeostasis.


Subject(s)
Diabetes, Gestational/immunology , Fetal Death/etiology , Receptor Activator of Nuclear Factor-kappa B/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Adipocytes/pathology , Animals , Cell Proliferation , Diabetes, Gestational/etiology , Diabetes, Gestational/metabolism , Diabetes, Gestational/pathology , Epithelial Cells/immunology , Female , Fetus/immunology , Fetus/metabolism , Fetus/pathology , Glucose/metabolism , Glucose Intolerance/genetics , Humans , Intra-Abdominal Fat/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Placenta/immunology , Placenta/pathology , Pregnancy , Receptor Activator of Nuclear Factor-kappa B/deficiency , Receptor Activator of Nuclear Factor-kappa B/genetics , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Transcription Factors/metabolism , AIRE Protein
4.
Blood ; 143(11): 1006-1017, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38142424

ABSTRACT

ABSTRACT: Systemic mastocytosis (SM) is defined by the expansion and accumulation of neoplastic mast cells (MCs) in the bone marrow (BM) and extracutaneous organs. Most patients harbor a somatic KIT D816V mutation, which leads to growth factor-independent KIT activation and accumulation of MC. Tumor necrosis factor α (TNF) is a proapoptotic and inflammatory cytokine that has been implicated in the clonal selection of neoplastic cells. We found that KIT D816V increases the expression and secretion of TNF. TNF expression in neoplastic MCs is reduced by KIT-targeting drugs. Similarly, knockdown of KIT or targeting the downstream signaling cascade of MAPK and NF-κB signaling reduced TNF expression levels. TNF reduces colony formation in human BM cells, whereas KIT D816V+ cells are less susceptible to the cytokine, potentially contributing to clonal selection. In line, knockout of TNF in neoplastic MC prolonged survival and reduced myelosuppression in a murine xenotransplantation model. Mechanistic studies revealed that the relative resistance of KIT D816V+ cells to TNF is mediated by the apoptosis-regulator BIRC5 (survivin). Expression of BIRC5 in neoplastic MC was confirmed by immunohistochemistry of samples from patients with SM. TNF serum levels are significantly elevated in patients with SM and high TNF levels were identified as a biomarker associated with inferior survival. We here characterized TNF as a KIT D816V-dependent cytokine that promotes clonal dominance. We propose TNF and apoptosis-associated proteins as potential therapeutic targets in SM.


Subject(s)
Mastocytosis, Systemic , Mastocytosis , Humans , Animals , Mice , Tumor Necrosis Factor-alpha , Survivin/genetics , Prognosis , Mastocytosis, Systemic/diagnosis , Mastocytosis, Systemic/genetics , Cytokines
5.
EMBO J ; 40(22): e108234, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34586646

ABSTRACT

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Subject(s)
DNA Methylation , Dermatitis/genetics , Epidermis/physiopathology , Nucleotidyltransferases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chromosome Aberrations , Cytosol/physiology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Dermatitis/immunology , Dermatitis/pathology , Humans , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic , Nucleotidyltransferases/genetics
6.
EMBO Rep ; 24(10): e57084, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37691494

ABSTRACT

Intestinal epithelial cells are covered by the brush border, which consists of densely packed microvilli. The Intermicrovillar Adhesion Complex (IMAC) links the microvilli and is required for proper brush border organization. Whether microvillus crosslinking is involved in the intestinal barrier function or colitis is currently unknown. We investigate the role of microvillus crosslinking in colitis in mice with deletion of the IMAC component CDHR5. Electron microscopy shows pronounced brush border defects in CDHR5-deficient mice. The defects result in severe mucosal damage after exposure to the colitis-inducing agent DSS. DSS increases the permeability of the mucus layer and brings bacteria in direct contact with the disorganized brush border of CDHR5-deficient mice. This correlates with bacterial invasion into the epithelial cell layer which precedes epithelial apoptosis and inflammation. Single-cell RNA sequencing data of patients with ulcerative colitis reveals downregulation of CDHR5 in enterocytes of diseased areas. Our results provide experimental evidence that a combination of microvillus crosslinking defects with increased permeability of the mucus layer sensitizes to inflammatory bowel disease.

7.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29118048

ABSTRACT

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Receptor Activator of Nuclear Factor-kappa B/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cell Respiration , Cells, Cultured , Energy Metabolism , Female , Gonadal Steroid Hormones/physiology , Homeostasis , Humans , Lung/metabolism , Lung Neoplasms/drug therapy , Male , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Respiratory Mucosa/metabolism
8.
Mol Cancer ; 23(1): 114, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811984

ABSTRACT

BACKGROUND: Prostate cancer develops through malignant transformation of the prostate epithelium in a stepwise, mutation-driven process. Although activator protein-1 transcription factors such as JUN have been implicated as potential oncogenic drivers, the molecular programs contributing to prostate cancer progression are not fully understood. METHODS: We analyzed JUN expression in clinical prostate cancer samples across different stages and investigated its functional role in a Pten-deficient mouse model. We performed histopathological examinations, transcriptomic analyses and explored the senescence-associated secretory phenotype in the tumor microenvironment. RESULTS: Elevated JUN levels characterized early-stage prostate cancer and predicted improved survival in human and murine samples. Immune-phenotyping of Pten-deficient prostates revealed high accumulation of tumor-infiltrating leukocytes, particularly innate immune cells, neutrophils and macrophages as well as high levels of STAT3 activation and IL-1ß production. Jun depletion in a Pten-deficient background prevented immune cell attraction which was accompanied by significant reduction of active STAT3 and IL-1ß and accelerated prostate tumor growth. Comparative transcriptome profiling of prostate epithelial cells revealed a senescence-associated gene signature, upregulation of pro-inflammatory processes involved in immune cell attraction and of chemokines such as IL-1ß, TNF-α, CCL3 and CCL8 in Pten-deficient prostates. Strikingly, JUN depletion reversed both the senescence-associated secretory phenotype and senescence-associated immune cell infiltration but had no impact on cell cycle arrest. As a result, JUN depletion in Pten-deficient prostates interfered with the senescence-associated immune clearance and accelerated tumor growth. CONCLUSIONS: Our results suggest that JUN acts as tumor-suppressor and decelerates the progression of prostate cancer by transcriptional regulation of senescence- and inflammation-associated genes. This study opens avenues for novel treatment strategies that could impede disease progression and improve patient outcomes.


Subject(s)
Disease Progression , PTEN Phosphohydrolase , Prostatic Neoplasms , Tumor Microenvironment , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Animals , Mice , Humans , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Tumor Microenvironment/immunology , Senescence-Associated Secretory Phenotype , Proto-Oncogene Proteins c-jun/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Gene Expression Profiling , Cellular Senescence/genetics , Disease Models, Animal
9.
Eur J Nucl Med Mol Imaging ; 51(9): 2833-2842, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38693454

ABSTRACT

BACKGROUND: Circulating-tumor DNA (ctDNA) and prostate-specific membrane antigen (PSMA) ligand positron-emission tomography (PET) enable minimal-invasive prostate cancer (PCa) detection and survival prognostication. The present study aims to compare their tumor discovery abilities and prognostic values. METHODS: One hundred thirty men with confirmed PCa (70.5 ± 8.0 years) who underwent [68Ga]Ga-PSMA-11 PET/CT (184.8 ± 19.7 MBq) imaging and plasma sample collection (March 2019-August 2021) were included. Plasma-extracted cell-free DNA was subjected to whole-genome-based ctDNA analysis. PSMA-positive tumor lesions were delineated and their quantitative parameters extracted. ctDNA and PSMA PET/CT discovery rates were compared, and the prognostic value for overall survival (OS) was evaluated. RESULTS: PSMA PET discovery rates according to castration status and PSA ranges did differ significantly (P = 0.013, P < 0.001), while ctDNA discovery rates did not (P = 0.311, P = 0.123). ctDNA discovery rates differed between localized and metastatic disease (P = 0.013). Correlations between ctDNA concentrations and PSMA-positive tumor volume (PSMA-TV) were significant in all (r = 0.42, P < 0.001) and castration-resistant (r = 0.65, P < 0.001), however not in hormone-sensitive patients (r = 0.15, P = 0.249). PSMA-TV and ctDNA levels were associated with survival outcomes in the Logrank (P < 0.0001, P < 0.0001) and multivariate Cox regression analysis (P = 0.0023, P < 0.0001). CONCLUSION: These findings suggest that PSMA PET imaging outperforms ctDNA analysis in detecting prostate cancer across the whole spectrum of disease, while both modalities are independently highly prognostic for survival outcomes.


Subject(s)
Circulating Tumor DNA , Edetic Acid , Gallium Isotopes , Gallium Radioisotopes , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/blood , Aged , Edetic Acid/analogs & derivatives , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Prognosis , Oligopeptides , Cross-Sectional Studies , Middle Aged
10.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33602823

ABSTRACT

Many cancers evade immune rejection by suppressing major histocompatibility class I (MHC-I) antigen processing and presentation (AgPP). Such cancers do not respond to immune checkpoint inhibitor therapies (ICIT) such as PD-1/PD-L1 [PD-(L)1] blockade. Certain chemotherapeutic drugs augment tumor control by PD-(L)1 inhibitors through potentiation of T-cell priming but whether and how chemotherapy enhances MHC-I-dependent cancer cell recognition by cytotoxic T cells (CTLs) is not entirely clear. We now show that the lysine acetyl transferases p300/CREB binding protein (CBP) control MHC-I AgPPM expression and neoantigen amounts in human cancers. Moreover, we found that two distinct DNA damaging drugs, the platinoid oxaliplatin and the topoisomerase inhibitor mitoxantrone, strongly up-regulate MHC-I AgPP in a manner dependent on activation of nuclear factor kappa B (NF-κB), p300/CBP, and other transcription factors, but independently of autocrine IFNγ signaling. Accordingly, NF-κB and p300 ablations prevent chemotherapy-induced MHC-I AgPP and abrogate rejection of low MHC-I-expressing tumors by reinvigorated CD8+ CTLs. Drugs like oxaliplatin and mitoxantrone may be used to overcome resistance to PD-(L)1 inhibitors in tumors that had "epigenetically down-regulated," but had not permanently lost MHC-I AgPP activity.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/immunology , Immune Checkpoint Inhibitors/pharmacology , NF-kappa B/metabolism , Neoplasms/drug therapy , p300-CBP Transcription Factors/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes , Cell Proliferation , Drug Therapy, Combination , Humans , Immunotherapy/methods , Mice , NF-kappa B/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Oxaliplatin/pharmacology , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/genetics
11.
Br J Haematol ; 202(5): 985-994, 2023 09.
Article in English | MEDLINE | ID: mdl-37357529

ABSTRACT

Anaplastic large-cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by the oncogenic anaplastic lymphoma kinase (ALK), accounting for approximately 15% of all paediatric non-Hodgkin lymphoma. Patients with central nervous system (CNS) relapse are particularly difficult to treat with a 3-year overall survival of 49% and a median survival of 23.5 months. The second-generation ALK inhibitor brigatinib shows superior penetration of the blood-brain barrier unlike the first-generation drug crizotinib and has shown promising results in ALK+ non-small-cell lung cancer. However, the benefits of brigatinib in treating aggressive paediatric ALK+ ALCL are largely unknown. We established a patient-derived xenograft (PDX) resource from ALK+ ALCL patients at or before CNS relapse serving as models to facilitate the development of future therapies. We show in vivo that brigatinib is effective in inducing the remission of PDX models of crizotinib-resistant (ALK C1156Y, TP53 loss) ALCL and furthermore that it is superior to crizotinib as a second-line approach to the treatment of a standard chemotherapy relapsed/refractory ALCL PDX pointing to brigatinib as a future therapeutic option.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lymphoma, Large-Cell, Anaplastic , Child , Humans , Anaplastic Lymphoma Kinase , Crizotinib/pharmacology , Crizotinib/therapeutic use , Receptor Protein-Tyrosine Kinases/therapeutic use , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/pathology , Heterografts , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use
12.
Eur J Nucl Med Mol Imaging ; 50(2): 546-558, 2023 01.
Article in English | MEDLINE | ID: mdl-36161512

ABSTRACT

PURPOSE: Head and neck squamous cell carcinomas (HNSCCs) are a molecularly, histologically, and clinically heterogeneous set of tumors originating from the mucosal epithelium of the oral cavity, pharynx, and larynx. This heterogeneous nature of HNSCC is one of the main contributing factors to the lack of prognostic markers for personalized treatment. The aim of this study was to develop and identify multi-omics markers capable of improved risk stratification in this highly heterogeneous patient population. METHODS: In this retrospective study, we approached this issue by establishing radiogenomics markers to identify high-risk individuals in a cohort of 127 HNSCC patients. Hybrid in vivo imaging and whole-exome sequencing were employed to identify quantitative imaging markers as well as genetic markers on pathway-level prognostic in HNSCC. We investigated the deductibility of the prognostic genetic markers using anatomical and metabolic imaging using positron emission tomography combined with computed tomography. Moreover, we used statistical and machine learning modeling to investigate whether a multi-omics approach can be used to derive prognostic markers for HNSCC. RESULTS: Radiogenomic analysis revealed a significant influence of genetic pathway alterations on imaging markers. A highly prognostic radiogenomic marker based on cellular senescence was identified. Furthermore, the radiogenomic biomarkers designed in this study vastly outperformed the prognostic value of markers derived from genetics and imaging alone. CONCLUSION: Using the identified markers, a clinically meaningful stratification of patients is possible, guiding the identification of high-risk patients and potentially aiding in the development of effective targeted therapies.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/diagnostic imaging , Squamous Cell Carcinoma of Head and Neck/genetics , Carcinoma, Squamous Cell/pathology , Retrospective Studies , Genetic Markers , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/genetics , Prognosis , Risk Assessment
13.
Oral Dis ; 2023 May 08.
Article in English | MEDLINE | ID: mdl-37154295

ABSTRACT

OBJECTIVES: The transforming growth factor-Beta (TGF-ß) pathway may be involved in the radioresistance of head and neck squamous cell carcinoma (HNSCC). This study analyzed TGF-ß receptor 1 (TGFBR1) expression in HNSCC patients and evaluated the antineoplastic and radiosensitizing effects of vactosertib, a novel TGFBR1 inhibitor, in vitro. MATERIALS AND METHODS: TGFBR1 expression was examined in HNSCC patients at the mRNA level in silico and the protein level by immunohistochemistry, including surgical specimens of primary tumors, matched lymph node metastasis, and recurrent disease. Furthermore, a novel small molecule TGFBR1 inhibitor was evaluated in HNSCC cell lines. Finally, an indirect coculture model using patient-derived cancer-associated fibroblasts was applied to mimic the tumor microenvironment. RESULTS: Patients with high TGFBR1 mRNA levels showed significantly worse overall survival in silico (OS, p = 0.024). At the protein level, an association between TGFBR1+ tumor and OS was observed for the subgroup with TGFBR1-stroma (p = 0.001). Those results prevailed in multivariable analysis. Inhibition of TGFBR1 showed antineoplastic effects in vitro. In combination with radiation, vactosertib showed synergistic effects. CONCLUSION: Our results indicate a high risk of death in tumorTGFBR1+ |stromaTGFBR1- expressing patients. In vitro data suggest a potential radiosensitizing effect of TGFBR1 inhibition by vactosertib.

14.
Eur Arch Otorhinolaryngol ; 280(1): 399-408, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36001136

ABSTRACT

Beta-catenin is known to be a vital component of the canonical Wnt signaling cascade, involved in the carcinogenesis of different solid tumors. We aimed to evaluate the effects of Beta-catenin inhibition in head and neck squamous cell carcinoma (HNSCC) in vitro. The small molecular compound MSAB was used to inhibit Wnt/Beta-catenin signaling in a human papillomavirus (HPV)-positive and HPV-negative cell line and its effects on cell proliferation, migration, colony formation, apoptosis, as well as radiosensitizing properties were assessed. Significant antineoplastic effects were observed in both cell lines. Interestingly, stronger anti-neoplastic and radiosensitizing effects were observed in the HPV-negative cell line, whereas stronger anti-migratory potential was detected in HPV-positive HNSCC cells. In conclusion, our findings suggest MSAB as a potential therapeutic agent for HNSCC. Further studies are warranted to unravel the mechanistic background of our findings.


Subject(s)
Head and Neck Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Head and Neck Neoplasms/drug therapy , beta Catenin/metabolism , Cell Line, Tumor
15.
Int J Mol Sci ; 25(1)2023 Dec 24.
Article in English | MEDLINE | ID: mdl-38203458

ABSTRACT

Her-2/neu-targeting therapy by passive application with trastuzumab is associated with acquired resistance and subsequent metastasis development, which is attributed to the upregulation of tumoral PD-L1 expression and the downregulation of Her-2/neu. We aimed to investigate this association, following active immunization with our recently constructed B-cell peptide-based Her-2/neu vaccines in both preclinical and clinical settings. Immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and combined positive score (CPS) were applied to evaluate Her-2/neu and PD-L1 expression using a murine syngeneic tumor model for Her-2/neu lung metastases and tumor biopsies from a gastric cancer patient with disease progression. A significant and concomitant reduction in Her-2/neu and the upregulation of PD-L1 expression was observed in vaccinated mice after 45 days, but not after 30 days, of metastases development. A significant increase in tumor-infiltrating B lymphocytes was observed at both time points. The downregulation of Her-2/neu and the upregulation of PD-L1 were observed in a patient's primary tumor at the disease progression time point but not prior to vaccination (Her-2/neu IHC: 3 to 0, FISH: 4.98 to 1.63; PD-L1 CPS: 0% to 5%). Our results further underline the need for combination therapy by targeting PD-L1 to prevent metastasis formation and immune evasion of Her-2/neu-positive and PD-L1-negative tumor cells.


Subject(s)
B7-H1 Antigen , Cancer Vaccines , Humans , Animals , Mice , Immune Evasion , In Situ Hybridization, Fluorescence , Oncogenes , Cancer Vaccines/therapeutic use , Disease Progression
16.
Wien Med Wochenschr ; 173(9-10): 232-238, 2023 Jun.
Article in German | MEDLINE | ID: mdl-36445601

ABSTRACT

Climate change is a threat to health and social security of billions of people. Health and quality of life are increasingly affected in many ways due to the climate crisis. Rising global temperatures are resulting in increasingly frequent and severe extreme weather events, contributing to further increase in inequality, discrimination, and injustice overall and in health care specifically. Furthermore, climatic conditions are also becoming increasingly suitable for the transmission of infectious diseases and their spread into new regions. Socio-economically disadvantaged regions with weak health infrastructure (e.g. Global South) will be hardly able to cope without specific support. The overriding imperative is to achieve reductions in greenhouse gas emissions from transport, energy and food production at global, national and regional levels to mitigate negative health impacts. The 2015 Paris Agreement must also be seen as crucial health agreement. Our paper aims to highlight ethical aspects of climate change in the health sector.


Subject(s)
Climate Change , Quality of Life , Humans
17.
Mol Cancer ; 21(1): 89, 2022 03 30.
Article in English | MEDLINE | ID: mdl-35354467

ABSTRACT

BACKGROUND: Frequent truncation mutations of the histone lysine N-methyltransferase KMT2C have been detected by whole exome sequencing studies in various cancers, including malignancies of the prostate. However, the biological consequences of these alterations in prostate cancer have not yet been elucidated. METHODS: To investigate the functional effects of these mutations, we deleted the C-terminal catalytic core motif of Kmt2c specifically in mouse prostate epithelium. We analysed the effect of Kmt2c SET domain deletion in a Pten-deficient PCa mouse model in vivo and of truncation mutations of KMT2C in a large number of prostate cancer patients. RESULTS: We show here for the first time that impaired KMT2C methyltransferase activity drives proliferation and PIN formation and, when combined with loss of the tumour suppressor PTEN, triggers loss of senescence, metastatic dissemination and dramatically reduces life expectancy. In Kmt2c-mutated tumours we show enrichment of proliferative MYC gene signatures and loss of expression of the cell cycle repressor p16INK4A. In addition, we observe a striking reduction in disease-free survival of patients with KMT2C-mutated prostate cancer. CONCLUSIONS: We identified truncating events of KMT2C as drivers of proliferation and PIN formation. Loss of PTEN and KMT2C in prostate cancer results in loss of senescence, metastatic dissemination and reduced life expectancy. Our data demonstrate the prognostic significance of KMT2C mutation status in prostate cancer patients. Inhibition of the MYC signalling axis may be a viable treatment option for patients with KMT2C truncations and therefore poor prognosis.


Subject(s)
Methyltransferases , Prostatic Neoplasms , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/physiology , Humans , Male , Methyltransferases/genetics , Mice , Mutation , Prostatic Neoplasms/metabolism , Exome Sequencing
18.
Blood ; 136(14): 1657-1669, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32573700

ABSTRACT

Anaplastic large cell lymphoma (ALCL) is a T-cell malignancy predominantly driven by a hyperactive anaplastic lymphoma kinase (ALK) fusion protein. ALK inhibitors, such as crizotinib, provide alternatives to standard chemotherapy with reduced toxicity and side effects. Children with lymphomas driven by nucleophosmin 1 (NPM1)-ALK fusion proteins achieved an objective response rate to ALK inhibition therapy of 54% to 90% in clinical trials; however, a subset of patients progressed within the first 3 months of treatment. The mechanism for the development of ALK inhibitor resistance is unknown. Through genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) activation and knockout screens in ALCL cell lines, combined with RNA sequencing data derived from ALK inhibitor-relapsed patient tumors, we show that resistance to ALK inhibition by crizotinib in ALCL can be driven by aberrant upregulation of interleukin 10 receptor subunit alpha (IL10RA). Elevated IL10RA expression rewires the STAT3 signaling pathway, bypassing otherwise critical phosphorylation by NPM1-ALK. IL-10RA expression does not correlate with response to standard chemotherapy in pediatric patients, suggesting that a combination of crizotinib and chemotherapy could prevent ALK inhibitor resistance-specific relapse.


Subject(s)
Antineoplastic Agents/pharmacology , Crizotinib/pharmacology , Drug Resistance, Neoplasm/genetics , Interleukin-10 Receptor alpha Subunit/genetics , Lymphoma, Large-Cell, Anaplastic/genetics , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Antineoplastic Agents/therapeutic use , CRISPR-Cas Systems , Cell Line , Crizotinib/therapeutic use , Dose-Response Relationship, Drug , Gene Editing , Gene Expression , Humans , Immunohistochemistry , Interleukin-10 Receptor alpha Subunit/metabolism , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Models, Biological , Nucleophosmin , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
19.
Int J Cancer ; 148(3): 731-747, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33034050

ABSTRACT

Androgen deprivation therapy (ADT) remains a key approach in the treatment of prostate cancer (PCa). However, PCa inevitably relapses and becomes ADT resistant. Besides androgens, there is evidence that thyroid hormone thyroxine (T4) and its active form 3,5,3'-triiodo-L-thyronine (T3) are involved in the progression of PCa. Epidemiologic evidences show a higher incidence of PCa in men with elevated thyroid hormone levels. The thyroid hormone binding protein µ-Crystallin (CRYM) mediates intracellular thyroid hormone action by sequestering T3 and blocks its binding to cognate receptors (TRα/TRß) in target tissues. We show in our study that low CRYM expression levels in PCa patients are associated with early biochemical recurrence and poor prognosis. Moreover, we found a disease stage-specific expression of CRYM in PCa. CRYM counteracted thyroid and androgen signaling and blocked intracellular choline uptake. CRYM inversely correlated with [18F]fluoromethylcholine (FMC) levels in positron emission tomography/magnetic resonance imaging of PCa patients. Our data suggest CRYM as a novel antagonist of T3- and androgen-mediated signaling in PCa. The role of CRYM could therefore be an essential control mechanism for the prevention of aggressive PCa growth.


Subject(s)
Crystallins/genetics , Crystallins/metabolism , Down-Regulation , Prostatic Neoplasms/pathology , Signal Transduction , Cell Line, Tumor , Choline/administration & dosage , Choline/analogs & derivatives , Cohort Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Metabolomics , Neoplasm Staging , PC-3 Cells , Positron Emission Tomography Computed Tomography , Prognosis , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Thyroid Hormone/genetics , Sequence Analysis, RNA , Tissue Array Analysis , Triiodothyronine/antagonists & inhibitors , Triiodothyronine/metabolism , mu-Crystallins
20.
Br J Haematol ; 192(2): 354-365, 2021 01.
Article in English | MEDLINE | ID: mdl-32880915

ABSTRACT

Burkitt lymphoma (BL) accounts for almost two-thirds of all B-cell non-Hodgkin lymphoma (B-NHL) in children and adolescents and is characterised by a MYC translocation and rapid cell turnover. Intensive chemotherapeutic regimens have been developed in recent decades, including the lymphomes malins B (LMB) protocol, which have resulted in a survival rate in excess of 90%. Recent clinical trials have focused on immunochemotherapy, with the addition of rituximab to chemotherapeutic backbones, showing encouraging results. Despite these advances, relapse and refractory disease occurs in up to 10% of patients and salvage options for these carry a dismal prognosis. Efforts to better understand the molecular and functional characteristics driving relapse and refractory disease may help improve this prognosis. This study has established a paediatric BL patient-derived xenograft (PDX) resource which captures and maintains tumour heterogeneity, may be used to better characterise tumours and identify cell populations responsible for therapy resistance.


Subject(s)
Burkitt Lymphoma/pathology , Animals , Burkitt Lymphoma/genetics , Burkitt Lymphoma/therapy , Child , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Heterografts/pathology , Humans , Male , Mice , Neoplasm Transplantation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL