Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 119(40): e2203307119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161914

ABSTRACT

Brown adipose tissue (BAT) is a highly specialized adipose tissue in its immobile location and size during the entire adulthood. In response to cold exposure and other ß3-adrenoreceptor stimuli, BAT commits energy consumption by nonshivering thermogenesis (NST). However, the molecular machinery in controlling the BAT mass in adults is unknown. Here, we show our surprising findings that the BAT mass and functions can be manipulated in adult animals by controlling BAT adipocyte differentiation in vivo. Platelet-derived growth factor receptor α (PDGFα) expressed in BAT progenitor cells served a signaling function to avert adipose progenitor differentiation. Genetic and pharmacological loss-of-function of PDGFRα eliminated the differentiation barrier and permitted progenitor cell differentiation to mature and functional BAT adipocytes. Consequently, an enlarged BAT mass (megaBAT) was created by PDGFRα inhibition owing to increases of brown adipocyte numbers. Under cold exposure, a microRNA-485 (miR-485) was identified as a master suppressor of the PDGFRα signaling, and delivery of miR-485 also produced megaBAT in adult animals. Noticeably, megaBAT markedly improved global metabolism, insulin sensitivity, high-fat-diet (HFD)-induced obesity, and diabetes by enhancing NST. Together, our findings demonstrate that the adult BAT mass can be increased by blocking the previously unprecedented inhibitory signaling for BAT progenitor cell differentiation. Thus, blocking the PDGFRα for the generation of megaBAT provides an attractive strategy for treating obesity and type 2 diabetes mellitus (T2DM).


Subject(s)
Adipocytes, Brown , Adipocytes , Adipogenesis , Adipose Tissue, Brown , MicroRNAs , Receptor, Platelet-Derived Growth Factor alpha , Adipocytes/cytology , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Diabetes Mellitus, Type 2/therapy , Energy Metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/therapy , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Thermogenesis/genetics
2.
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
3.
Neurology ; 95(24): e3163-e3179, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33144514

ABSTRACT

OBJECTIVE: To test the hypothesis that monogenic neuropathies such as Charcot-Marie-Tooth disease (CMT) contribute to frequent but often unexplained neuropathies in the elderly, we performed genetic analysis of 230 patients with unexplained axonal neuropathies and disease onset ≥35 years. METHODS: We recruited patients, collected clinical data, and conducted whole-exome sequencing (WES; n = 126) and MME single-gene sequencing (n = 104). We further queried WES repositories for MME variants and measured blood levels of the MME-encoded protein neprilysin. RESULTS: In the WES cohort, the overall detection rate for assumed disease-causing variants in genes for CMT or other conditions associated with neuropathies was 18.3% (familial cases 26.4%, apparently sporadic cases 12.3%). MME was most frequently involved and accounted for 34.8% of genetically solved cases. The relevance of MME for late-onset neuropathies was further supported by detection of a comparable proportion of cases in an independent patient sample, preponderance of MME variants among patients compared to population frequencies, retrieval of additional late-onset neuropathy patients with MME variants from WES repositories, and low neprilysin levels in patients' blood samples. Transmission of MME variants was often consistent with an incompletely penetrant autosomal-dominant trait and less frequently with autosomal-recessive inheritance. CONCLUSIONS: A detectable fraction of unexplained late-onset axonal neuropathies is genetically determined, by variants in either CMT genes or genes involved in other conditions that affect the peripheral nerves and can mimic a CMT phenotype. MME variants can act as completely penetrant recessive alleles but also confer dominantly inherited susceptibility to axonal neuropathies in an aging population.


Subject(s)
Aging , Hereditary Sensory and Motor Neuropathy/genetics , Neprilysin/genetics , Age of Onset , Aged , Aging/blood , Charcot-Marie-Tooth Disease/blood , Charcot-Marie-Tooth Disease/genetics , Female , Genetic Predisposition to Disease/genetics , Hereditary Sensory and Motor Neuropathy/blood , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Neprilysin/blood , Exome Sequencing
5.
Pathogens ; 8(4)2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31618932

ABSTRACT

Ebola virus (EBOV) infection can cause severe and frequently fatal disease in human patients. The EBOV glycoprotein (GP) mediates viral entry into host cells. For this, GP depends on priming by the pH-dependent endolysosomal cysteine proteases cathepsin B (CatB) and, to a lesser degree, cathepsin L (CatL), at least in most cell culture systems. However, there is limited information on whether and how EBOV-GP can acquire resistance to CatB/L inhibitors. Here, we addressed this question using replication-competent vesicular stomatitis virus bearing EBOV-GP. Five passages of this virus in the presence of the CatB/CatL inhibitor MDL28170 were sufficient to select resistant viral variants and sequencing revealed that all GP sequences contained a V37A mutation, which, in the context of native GP, is located in the base of the GP surface unit. In addition, some GP sequences harbored mutation S195R in the receptor-binding domain. Finally, mutational analysis demonstrated that V37A but not S195R conferred resistance against MDL28170 and other CatB/CatL inhibitors. Collectively, a single amino acid substitution in GP is sufficient to confer resistance against CatB/CatL inhibitors, suggesting that usage of CatB/CatL inhibitors for antiviral therapy may rapidly select for resistant viral variants.

6.
Sci Rep ; 9(1): 4096, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858407

ABSTRACT

Melanoma is a leading cause of high mortality that frequently spreads to the brain and is associated with deterioration in quality and quantity of life. Treatment opportunities have been restricted until now and new therapy options are urgently required. Our focus was to reveal the potential heterogeneity of melanoma brain metastasis. We succeeded to establish a brain melanoma metastasis cell line, namely MUG-Mel1 and two resulting clones D5 and C8 by morphological variety, differences in lipidome, growth behavior, surface, and stem cell markers. Mutation analysis by next-generation sequencing, copy number profiling, and cytogenetics demonstrated the different genetic profile of MUG-Mel1 and clones. Tumorigenicity was unsuccessfully tested in various mouse systems and finally established in a zebra fish model. As innovative treatment option, with high potential to pass the blood-brain barrier a peptide isolated from lactoferricin was studied in potential toxicity. Brain metastases are a major clinical challenge, therefore the development of relevant in vitro and in vivo models derived from brain melanoma metastases provides valuable information about tumor biology and offers great potential to screen for new innovative therapies.


Subject(s)
Brain Neoplasms/secondary , Clone Cells/pathology , Melanoma/pathology , Animals , Brain Neoplasms/ultrastructure , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Female , Gene Dosage , Humans , Inhibitory Concentration 50 , Lipids/analysis , Male , Melanoma/ultrastructure , Mice, Nude , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Peptides/pharmacology , Zebrafish
7.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333314

ABSTRACT

Molecular mechanisms underlying the cancer stroma in metastasis need further exploration. Here, we discovered that cancer-associated fibroblasts (CAFs) produced high levels of IL-33 that acted on tumor-associated macrophages (TAMs), causing them to undergo the M1 to M2 transition. Genomic profiling of metastasis-related genes in the IL-33-stimulated TAMs showed a >200-fold increase of MMP9. Signaling analysis demonstrated the IL-33-ST2-NF-κB-MMP9-laminin pathway that governed tumor stroma-mediated metastasis. In mouse and human fibroblast-rich pancreatic cancers, genetic deletion of IL-33, ST2, or MMP9 markedly blocked metastasis. Pharmacological inhibition of NF-κB and MMP9 also blocked cancer metastasis. Deletion of IL-33, ST2, or MMP9 restored laminin, a key basement membrane component associated with tumor microvessels. Together, our data provide mechanistic insights on the IL-33-NF-κB-MMP9-laminin axis that mediates the CAF-TAM-committed cancer metastasis. Thus, targeting the CAF-TAM-vessel axis provides an outstanding therapeutic opportunity for cancer treatment.


Subject(s)
Interleukin-33/metabolism , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Communication/immunology , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Interleukin-33/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasms/pathology , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
8.
J Exp Med ; 215(2): 611-626, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29305395

ABSTRACT

Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)-fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.


Subject(s)
Adipose Tissue/blood supply , Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Metabolic Diseases/therapy , Vascular Endothelial Growth Factor Receptor-1/deficiency , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Obesity/etiology , Obesity/metabolism , Obesity/therapy , Thermogenesis , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
9.
Nat Commun ; 8(1): 2079, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233981

ABSTRACT

Understanding the molecular mechanisms regulating beige adipocyte formation may lead to the development of new therapies to combat obesity. Here, we report a miRNA-based autocrine regulatory pathway that controls differentiation of preadipocytes into beige adipocytes. We identify miR-327 as one of the most downregulated miRNAs targeting growth factors in the stromal-vascular fraction (SVF) under conditions that promote white adipose tissue (WAT) browning in mice. Gain- and loss-of-function experiments reveal that miR-327 targets FGF10 to prevent beige adipocyte differentiation. Pharmacological and physiological ß-adrenergic stimulation upregulates FGF10 levels and promotes preadipocyte differentiation into beige adipocytes. In vivo local delivery of miR-327 to WATs significantly compromises the beige phenotype and thermogenesis. Contrarily, systemic inhibition of miR-327 in mice induces browning and increases whole-body metabolic rate under thermoneutral conditions. Our data provide mechanistic insight into an autocrine regulatory signaling loop that regulates beige adipocyte formation and suggests that the miR-327-FGF10-FGFR2 signaling axis may be a therapeutic targets for treatment of obesity and metabolic diseases.


Subject(s)
Adipose Tissue, Beige/physiology , Adipose Tissue, White/physiology , Autocrine Communication/genetics , Fibroblast Growth Factor 10/genetics , MicroRNAs/metabolism , Adipocytes/physiology , Adipose Tissue, Beige/cytology , Adipose Tissue, Beige/drug effects , Adipose Tissue, White/cytology , Adipose Tissue, White/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Down-Regulation , Energy Metabolism/physiology , Female , Fibroblast Growth Factor 10/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , MicroRNAs/genetics , Models, Animal , Obesity/drug therapy , Obesity/metabolism , RNA, Small Interfering/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/physiology
10.
Methods Mol Biol ; 1551: 87-112, 2017.
Article in English | MEDLINE | ID: mdl-28138842

ABSTRACT

The Human Major Histocompatibility Complex (MHC) is a highly polymorphic region full of immunoregulatory genes. The MHC codes for the human leukocyte antigens (HLA), proteins that present on the cellular surface and that are involved in self-non-self recognition. For matching donors and recipients for organ and stem-cell transplants it is important to know an individual's HLA haplotype determinable in this region. Now, as next-generation sequencing (NGS) platforms mature and become more and more accepted as a standard method, NGS applications have spread from research laboratories to the clinic, where they provide valid genetic insights. Here, we describe a cost-effective microarray-based sequence capture, enrichment, and NGS sequencing approach to characterize MHC haplotypes. Using this approach, ~4 MB of MHC sequence for four DNA samples (donor, recipient and the parents of the recipient) were sequenced in parallel in one NGS instrument run. We complemented this approach using microarray-based genome-wide SNP analysis. Taken together, the use of recently developed tools and protocols for sequence capture and massively parallel sequencing allows for detailed MHC analysis and donor-recipient matching.


Subject(s)
Major Histocompatibility Complex/genetics , Genotype , HLA Antigens/genetics , Haplotypes/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods
11.
JCI Insight ; 2(4): e89044, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239649

ABSTRACT

Visceral fat is considered the genuine and harmful white adipose tissue (WAT) that is associated to development of metabolic disorders, cardiovascular disease, and cancer. Here, we present a new concept to turn the harmful visceral fat into a beneficial energy consumption depot, which is beneficial for improvement of metabolic dysfunctions in obese mice. We show that low temperature-dependent browning of visceral fat caused decreased adipose weight, total body weight, and body mass index, despite increased food intake. In high-fat diet-fed mice, low temperature exposure improved browning of visceral fat, global metabolism via nonshivering thermogenesis, insulin sensitivity, and hepatic steatosis. Genome-wide expression profiling showed upregulation of WAT browning-related genes including Cidea and Dio2. Conversely, Prdm16 was unchanged in healthy mice or was downregulated in obese mice. Surgical removal of visceral fat and genetic knockdown of UCP1 in epididymal fat largely ablated low temperature-increased global thermogenesis and resulted in the death of most mice. Thus, browning of visceral fat may be a compensatory heating mechanism that could provide a novel therapeutic strategy for treating visceral fat-associated obesity and diabetes.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cold Temperature , Energy Metabolism , Intra-Abdominal Fat/metabolism , Thermogenesis , Adiponectin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Body Temperature , Body Weight , DNA-Binding Proteins/genetics , Diet, High-Fat , Eating , Fatty Liver , Gene Knockdown Techniques , Insulin Resistance , Iodide Peroxidase/genetics , Leptin/metabolism , Mice , Mice, Obese , Organ Size , Transcription Factors/genetics , Uncoupling Protein 1/genetics , Up-Regulation , Iodothyronine Deiodinase Type II
12.
Proc Natl Acad Sci U S A ; 113(38): E5618-27, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27608497

ABSTRACT

Vascular pericytes, an important cellular component in the tumor microenvironment, are often associated with tumor vasculatures, and their functions in cancer invasion and metastasis are poorly understood. Here we show that PDGF-BB induces pericyte-fibroblast transition (PFT), which significantly contributes to tumor invasion and metastasis. Gain- and loss-of-function experiments demonstrate that PDGF-BB-PDGFRß signaling promotes PFT both in vitro and in in vivo tumors. Genome-wide expression analysis indicates that PDGF-BB-activated pericytes acquire mesenchymal progenitor features. Pharmacological inhibition and genetic deletion of PDGFRß ablate the PDGF-BB-induced PFT. Genetic tracing of pericytes with two independent mouse strains, TN-AP-CreERT2:R26R-tdTomato and NG2-CreERT2:R26R-tdTomato, shows that PFT cells gain stromal fibroblast and myofibroblast markers in tumors. Importantly, coimplantation of PFT cells with less-invasive tumor cells in mice markedly promotes tumor dissemination and invasion, leading to an increased number of circulating tumor cells and metastasis. Our findings reveal a mechanism of vascular pericytes in PDGF-BB-promoted cancer invasion and metastasis by inducing PFT, and thus targeting PFT may offer a new treatment option of cancer metastasis.


Subject(s)
Carcinoma, Renal Cell/genetics , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Animals , Becaplermin , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
13.
Nat Commun ; 7: 12680, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27580750

ABSTRACT

The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the 'off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Indoles/therapeutic use , Liver/blood supply , Liver/pathology , Neoplasms/pathology , Pyrroles/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Female , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Sunitinib , Vascular Endothelial Growth Factor A/genetics
14.
Nat Commun ; 7: 12152, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27492130

ABSTRACT

Cold- and ß3-adrenoceptor agonist-induced sympathetic activation leads to angiogenesis and UCP1-dependent thermogenesis in mouse brown and white adipose tissues. Here we show that endothelial production of PDGF-CC during white adipose tissue (WAT) angiogenesis regulates WAT browning. We find that genetic deletion of endothelial VEGFR2, knockout of the Pdgf-c gene or pharmacological blockade of PDGFR-α impair the WAT-beige transition. We further show that PDGF-CC stimulation upregulates UCP1 expression and acquisition of a beige phenotype in differentiated mouse WAT-PDGFR-α(+) progenitor cells, as well as in human WAT-PDGFR-α(+) adipocytes, supporting the physiological relevance of our findings. Our data reveal a paracrine mechanism by which angiogenic endothelial cells modulate adipocyte metabolism, which may provide new targets for the treatment of obesity and related metabolic diseases.


Subject(s)
Adipose Tissue, Beige/blood supply , Adipose Tissue, Beige/physiology , Endothelial Cells/metabolism , Lymphokines/metabolism , Neovascularization, Physiologic , Platelet-Derived Growth Factor/metabolism , Thermogenesis , Animals , Diet, High-Fat , Dioxoles/pharmacology , Endothelial Cells/drug effects , Female , Gene Deletion , Humans , Intra-Abdominal Fat/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Neovascularization, Physiologic/drug effects , Phenotype , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Adrenergic/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
J Cancer ; 6(10): 984-9, 2015.
Article in English | MEDLINE | ID: mdl-26366211

ABSTRACT

BACKGROUND: Chordoma is a rare primary malignant bone tumour. Treatment options are mainly restricted to surgical excision, since chordomas are largely resistant to conventional ionising radiation and chemotherapy. Thus, there is a strong need to gain more thorough insights into the molecular biology and genetics of chordoma to allow for the development of new therapeutic options. We performed an ultra-deep sequencing analysis to find novel mutations in cancer associated genes in chordomas to date unseen with Sanger sequencing. MATERIAL AND METHODS: Nine chordomas (skull base (n=3), mobile spine (n=4), and sacrum/coccyx (n=2) were screened for mutations in 48 cancer genes using the Hot Spot Cancer Panel (Illumina). All putative mutations were compared against multiple databases (e.g. NCBI, COSMIC, PolyPhen, EGB, SIFT) and published Copy Number Variation (CNV) data for chordoma. RESULTS: Our results showed mutations with a frequency above 5% in tumorsuppressor- and onco-genes, revealing new possible driver genes for chordomas. We detected three different variants accounting for 11 point mutations in three cancer associated genes (KIT, KDR and TP53). None of the detected mutations was found in all samples investigated. However, all genes affected interact or are connected in pathway analysis. There were no correlations to already reported CNVs in the samples analysed. CONCLUSIONS: We identified mutations in the associated genes KIT, KDR, and TP53. These mutations have been described previously and have been predicted to be tolerated. Further results on a larger series are warranted. The driver mechanisms of chordoma still have to be identified.

16.
PLoS One ; 10(3): e0119576, 2015.
Article in English | MEDLINE | ID: mdl-25751574

ABSTRACT

BACKGROUND: In recent years various studies showed, that hepatitis E virus (HEV) is a growing public health problem in many developed countries. Therefore, HEV infections might bear a transmission risk by blood transfusions. The clinical relevance still requires further investigations. The aim of this study was to provide an overview of acute HEV infections in Upper Austrian blood donors as well as a risk estimation of this transfusion-related infection. METHODS AND FINDINGS: A total of 58,915 blood donors were tested for HEV RNA using a commercial HEV RT-PCR Kit. 7 of these donors (0.01%) were PCR-positive with normal laboratory parameters in absence of clinical signs of hepatitis. Viral load determined by quantitative real-time PCR showed a HEV nucleic acid concentration of 2,217 293,635 IU/ml. At follow-up testing (2-11 weeks after donation) all blood donors had negative HEV RNA results. Additionally, genotyping was performed by amplification and sequencing of the ORF1 or ORF2 region of the HEV genome. All HEV RNA positive donor samples revealed a genotype 3 isolate. For the antibody screening, anti-HEV IgM and IgG were detected by ELISA. Follow up serological testing revealed that no donor was seropositive for HEV IgM or IgG antibodies at time of donation. Moreover, we verified the prevalence of anti-HEV IgG in 1,203 of the HEV RNA negative tested blood donors. Overall 13.55% showed positive results for anti-HEV IgG. CONCLUSIONS: In the presented study, we investigated HEV infections in blood donations of Upper Austria over 1 year. We concluded that 1 out of 8,416 blood donations is HEV RNA positive. Seroprevalence of anti HEV IgG results in an age-related increase of 13.55%. Therefore, based on this data, we recommend HEV-PCR screening to prevent transmission of hepatitis E virus by transfusion.


Subject(s)
Blood Donors , Hepatitis E virus/immunology , Hepatitis E/epidemiology , Adult , Antibodies, Viral/blood , Austria/epidemiology , Female , Hepatitis E/blood , Hepatitis E/immunology , Humans , Immunoglobulin G/blood , Incidence , Male , Middle Aged , Seroepidemiologic Studies , Young Adult
17.
Mol Cancer ; 14: 4, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25608569

ABSTRACT

BACKGROUND: Lung cancer is one of the most frequent cancer types and the leading cause of cancer death worldwide. Cisplatin is a widely used chemotherapeutic for non-small cell lung carcinoma (NSCLC), however, its positive effects are diminished under hypoxia. We wanted to determine if co-treatment with cisplatin and histone deacetalyse (HDAC) inhibitor panobinostat can reduce hypoxia-induced cisplatin resistance in NSCLC cells, and to elucidate mechanism involved. METHODS: Expression status of different HDACS was determined in two cell lines and in tumor tissue from 20 patients. Cells were treated with cisplatin, panobinostat, or with combination of both under normoxic and hypoxic (1% O(2)) conditions. Cell cycle, viability, acetylation of histones, and activation of apoptosis were determined. HIF-1α stability and its interaction with HDAC4 were analyzed. RESULTS: Most class I and II HDACs were expressed in NSCLC cells and tumor samples. Co-treatment of tumor cells with cisplatin and panobinostat decreased cell viability and increased apoptosis more efficiently than in primary, non-malignant bronchial epithelial cells. Co-treatment induced apoptosis by causing chromatin fragmentation, activation of caspases-3 and 7 and PARP cleavage. Toxic effects were more pronounced under hypoxic conditions. Co-treatment resulted in destabilization and degradation of HIF-1α and HDAC4, a protein responsible for acetylation and de/stabilization of HIF-1α. Direct interaction between HDAC4 and HIF-1α proteins in H23 cells was detected. CONCLUSIONS: Here we show that hypoxia-induced cisplatin resistance can be overcome by combining cisplatin with panobinostat, a potent HDAC inhibitor. These findings may contribute to the development of a new therapeutic strategy for NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Hydroxamic Acids/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Indoles/pharmacology , Lung Neoplasms/metabolism , Acetylation , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Drug Synergism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Lung Neoplasms/genetics , Panobinostat , Spheroids, Cellular/drug effects , Tumor Cells, Cultured
18.
Horm Mol Biol Clin Investig ; 19(1): 5-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25390012

ABSTRACT

The number of obese and overweight individuals is globally rising, and obesity-associated disorders such as type 2 diabetes, cardiovascular disease and certain types of cancer are among the most common causes of death. While white adipose tissue is the key player in the storage of energy, active brown adipose tissue expends energy due to its thermogenic capacity. Expanding and activating brown adipose tissue using pharmacological approaches therefore might offer an attractive possibility for therapeutic intervention to counteract obesity and its consequences for metabolic health.


Subject(s)
Adipose Tissue, Brown/metabolism , Neovascularization, Physiologic , Thermogenesis/physiology , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Adipose Tissue, White/physiopathology , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Obesity/metabolism , Obesity/physiopathology , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Uncoupling Protein 1
19.
Proc Natl Acad Sci U S A ; 111(41): 14906-11, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25271320

ABSTRACT

Mechanisms underlying age-related obesity and insulin resistance are generally unknown. Here, we report age-related adipose vascular changes markedly modulated fat mass, adipocyte functions, blood lipid composition, and insulin sensitivity. Notably, VEGF expression levels in various white adipose tissues (WATs) underwent changes uninterruptedly in different age populations. Anti-VEGF and anti- VEGF receptor 2 treatment in different age populations showed marked variations of vascular regression, with midaged mice exhibiting modest sensitivity. Interestingly, anti-VEGF treatment produced opposing effects on WAT adipocyte sizes in different age populations and affected vascular density and adipocyte sizes in brown adipose tissue. Consistent with changes of vasculatures and adipocyte sizes, anti-VEGF treatment increased insulin sensitivity in young and old mice but had no effects in the midaged group. Surprisingly, anti-VEGF treatment significantly improved insulin sensitivity in midaged obese mice fed a high-fat diet. Our findings demonstrate that adipose vasculatures show differential responses to anti-VEGF treatment in various age populations and have therapeutic implications for treatment of obesity and diabetes with anti-VEGF-based antiangiogenic drugs.


Subject(s)
Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/blood supply , Adipose Tissue, White/physiopathology , Aging/pathology , Insulin Resistance , Vascular Endothelial Growth Factor A/metabolism , Adipocytes/pathology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Animals , Cell Size , Cell Survival/drug effects , Female , Lipids/blood , Mice, Inbred C57BL , Obesity/blood , Obesity/pathology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
EMBO J ; 32(24): 3176-91, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24240174

ABSTRACT

The histone deacetylases HDAC1 and HDAC2 remove acetyl moieties from lysine residues of histones and other proteins and are important regulators of gene expression. By deleting different combinations of Hdac1 and Hdac2 alleles in the epidermis, we reveal a dosage-dependent effect of HDAC1/HDAC2 activity on epidermal proliferation and differentiation. Conditional ablation of either HDAC1 or HDAC2 in the epidermis leads to no obvious phenotype due to compensation by the upregulated paralogue. Strikingly, deletion of a single Hdac2 allele in HDAC1 knockout mice results in severe epidermal defects, including alopecia, hyperkeratosis, hyperproliferation and spontaneous tumour formation. These mice display impaired Sin3A co-repressor complex function, increased levels of c-Myc protein, p53 expression and apoptosis in hair follicles (HFs) and misregulation of HF bulge stem cells. Surprisingly, ablation of HDAC1 but not HDAC2 in a skin tumour model leads to accelerated tumour development. Our data reveal a crucial function of HDAC1/HDAC2 in the control of lineage specificity and a novel role of HDAC1 as a tumour suppressor in the epidermis.


Subject(s)
Epidermis/growth & development , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Skin Neoplasms/genetics , Alopecia/genetics , Animals , Apoptosis/genetics , Cell Lineage , Co-Repressor Proteins , Disease Models, Animal , Epidermis/enzymology , Epidermis/pathology , Gene Expression Regulation , Genes, Tumor Suppressor , Genes, p53 , Hair Follicle/pathology , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Keratosis/genetics , Keratosis/pathology , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...