11 results on '"Hänisch B"'
Search Results
2. Replication of Functional Serotonin Receptor Type 3A and B Variants in Bipolar Affective Disorder: A European Multicenter Study
- Author
-
Hammer, C, Cichon, S, Mühleisen, T W, Haenisch, B, Degenhardt, F, Breuer, R, Witt, S H, Strohmaier, J, Oruc, L, Rivas, F, Babadjanova, G, Grigoroiu-Serbanescu, M, Röth, R, Rappold, G, Rietschel, M, Nöthen, M M, Niesler, B, Mattheisen, Manuel, and Hauser, J
- Subjects
association ,bipolar ,BPAD ,HTR3 ,HTR3B ,5-HT3 - Abstract
Serotonin type 3 receptors (\(5-HT_3\)) are involved in learning, cognition and emotion, and have been implicated in various psychiatric phenotypes. However, their contribution to the pathomechanism of these disorders remains elusive. Three single nucleotide polymorphisms (SNPs) in the \(HTR3A\) and \(HTR3B\) genes (rs1062613, rs1176744 and rs3831455) have been associated with bipolar affective disorder (BPAD) in pilot studies, and all of them are of functional relevance. We performed a European multicenter study to confirm previous results and provide further evidence for the relevance of these SNPs to the etiology of neuropsychiatric disorders. This involved analysis of the distribution of the three SNPs among 1804 BPAD cases and 2407 healthy controls. A meta-analysis revealed a pooled odds ratio of 0.881 (P=0.009, 95% confidence intervals=0.802–0.968) for the non-synonymous functional SNP \(HTR3B\) p.Y129S (rs1176744), thereby confirming previous findings. In line with this, the three genome-wide association study samples BOMA (Bonn-Mannheim)-BPAD, WTCCC (Wellcome Trust Case Control Consortium)-BPAD and GAIN (Genetic Association Information Network)-BPAD, including >3500 patients and 5200 controls in total, showed an overrepresentation of the p.Y129 in patients. Remarkably, the meta-analysis revealed a P-value of 0.048 (OR=0.934, fixed effect model). We also performed expression analyses to gain further insights into the distribution of \(HTR3A\) and \(HTR3B\) mRNA in the human brain. \(HTR3A\) and \(HTR3B\) were detected in all investigated brain tissues with the exception of the cerebellum, and large differences in the A:B subunit ratio were observed. Interestingly, expression of the B subunit was most prominent in the brain stem, amygdalae and frontal cortex, regions of relevance to psychiatric disorders. In conclusion, the present study provides further evidence for the presence of impaired \(5-HT_3\) receptor function in BPAD.
- Published
- 2012
- Full Text
- View/download PDF
3. Systematic annotation of orphan RNAs reveals blood-accessible molecular barcodes of cancer identity and cancer-emergent oncogenic drivers.
- Author
-
Wang J, Suh JM, Woo BJ, Navickas A, Garcia K, Yin K, Fish L, Cavazos T, Hänisch B, Markett D, Yu S, Hirst G, Brown-Swigart L, Esserman LJ, van 't Veer LJ, and Goodarzi H
- Abstract
From extrachromosomal DNA to neo-peptides, the broad reprogramming of the cancer genome leads to the emergence of molecules that are specific to the cancer state. We recently described orphan non-coding RNAs (oncRNAs) as a class of cancer-specific small RNAs with the potential to play functional roles in breast cancer progression
1 . Here, we report a systematic and comprehensive search to identify, annotate, and characterize cancer-emergent oncRNAs across 32 tumor types. We also leverage large-scale in vivo genetic screens in xenografted mice to functionally identify driver oncRNAs in multiple tumor types. We have not only discovered a large repertoire of oncRNAs, but also found that their presence and absence represent a digital molecular barcode that faithfully captures the types and subtypes of cancer. Importantly, we discovered that this molecular barcode is partially accessible from the cell-free space as some oncRNAs are secreted by cancer cells. In a large retrospective study across 192 breast cancer patients, we showed that oncRNAs can be reliably detected in the blood and that changes in the cell-free oncRNA burden captures both short-term and long-term clinical outcomes upon completion of a neoadjuvant chemotherapy regimen. Together, our findings establish oncRNAs as an emergent class of cancer-specific non-coding RNAs with potential roles in tumor progression and clinical utility in liquid biopsies and disease monitoring., Competing Interests: Disclosure of Potential Competing Interest H.G. is a co-founder and shareholder of Exai Bio. J.W., L.F., and T.C. are employees and shareholders of Exai Bio. L.J.E. reports funding from Merck & Co.; participation on an advisory board for Blue Cross Blue Shield; and personal fees from UpToDate. L.J.v.V. is a founding advisor and shareholder of Exai BIo; part-time employee and owns stock in Agendia. All other authors declare no competing interests.- Published
- 2024
- Full Text
- View/download PDF
4. Cerebral chemoarchitecture shares organizational traits with brain structure and function.
- Author
-
Hänisch B, Hansen JY, Bernhardt BC, Eickhoff SB, Dukart J, Misic B, and Valk SL
- Subjects
- Humans, Brain Mapping, Positron-Emission Tomography, Diffusion Magnetic Resonance Imaging, Magnetic Resonance Imaging methods, Brain diagnostic imaging
- Abstract
Chemoarchitecture, the heterogeneous distribution of neurotransmitter transporter and receptor molecules, is a relevant component of structure-function relationships in the human brain. Here, we studied the organization of the receptome, a measure of interareal chemoarchitectural similarity, derived from positron-emission tomography imaging studies of 19 different neurotransmitter transporters and receptors. Nonlinear dimensionality reduction revealed three main spatial gradients of cortical chemoarchitectural similarity - a centro-temporal gradient, an occipito-frontal gradient, and a temporo-occipital gradient. In subcortical nuclei, chemoarchitectural similarity distinguished functional communities and delineated a striato-thalamic axis. Overall, the cortical receptome shared key organizational traits with functional and structural brain anatomy, with node-level correspondence to functional, microstructural, and diffusion MRI-based measures decreasing along a primary-to-transmodal axis. Relative to primary and paralimbic regions, unimodal and heteromodal regions showed higher receptomic diversification, possibly supporting functional flexibility., Competing Interests: BH, JH, BB, SE, JD, BM, SV No competing interests declared, (© 2023, Hänisch et al.)
- Published
- 2023
- Full Text
- View/download PDF
5. Stage-specific transcription activator ESB1 regulates monoallelic antigen expression in Trypanosoma brucei.
- Author
-
López-Escobar L, Hänisch B, Halliday C, Ishii M, Akiyoshi B, Dean S, Sunter JD, Wheeler RJ, and Gull K
- Subjects
- Antigenic Variation genetics, Membrane Glycoproteins metabolism, RNA Polymerase I genetics, RNA Polymerase I metabolism, Transcription Factors genetics, Variant Surface Glycoproteins, Trypanosoma metabolism, Trypanosoma brucei brucei
- Abstract
Variant surface glycoprotein (VSG) coats bloodstream form Trypanosoma brucei parasites, and monoallelic VSG expression underpins the antigenic variation necessary for pathogenicity. One of thousands of VSG genes is transcribed by RNA polymerase I in a singular nuclear structure called the expression site body (ESB), but how monoallelic VSG transcription is achieved remains unclear. Using a localization screen of 153 proteins we found one, ESB-specific protein 1 (ESB1), that localized only to the ESB and is expressed only in VSG-expressing life cycle stages. ESB1 associates with DNA near the active VSG promoter and is necessary for VSG expression, with overexpression activating inactive VSG promoters. Mechanistically, ESB1 is necessary for recruitment of a subset of ESB components, including RNA polymerase I, revealing that the ESB has separately assembled subdomains. Because many trypanosomatid parasites have divergent ESB1 orthologues yet do not undergo antigenic variation, ESB1 probably represents an important class of transcription regulators., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
6. A prometastatic splicing program regulated by SNRPA1 interactions with structured RNA elements.
- Author
-
Fish L, Khoroshkin M, Navickas A, Garcia K, Culbertson B, Hänisch B, Zhang S, Nguyen HCB, Soto LM, Dermit M, Mardakheh FK, Molina H, Alarcón C, Najafabadi HS, and Goodarzi H
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Algorithms, Animals, Binding Sites, Breast Neoplasms genetics, Cell Line, Tumor, Disease Progression, Exons, Gene Knockdown Techniques, Humans, Lung Neoplasms secondary, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Invasiveness, Neoplasm Transplantation, Nucleic Acid Conformation, Plectin genetics, Protein Binding, RNA Interference, RNA, Small Nuclear chemistry, RNA, Small Nuclear metabolism, RNA-Seq, Ribonucleoprotein, U2 Small Nuclear genetics, Software, Spliceosomes metabolism, Tumor Suppressor Proteins genetics, Alternative Splicing, Breast Neoplasms pathology, Neoplasm Metastasis genetics, RNA genetics, RNA metabolism, Ribonucleoprotein, U2 Small Nuclear metabolism
- Abstract
Aberrant alternative splicing is a hallmark of cancer, yet the underlying regulatory programs that control this process remain largely unknown. Here, we report a systematic effort to decipher the RNA structural code that shapes pathological splicing during breast cancer metastasis. We discovered a previously unknown structural splicing enhancer that is enriched near cassette exons with increased inclusion in highly metastatic cells. We show that the spliceosomal protein small nuclear ribonucleoprotein polypeptide A' (SNRPA1) interacts with these enhancers to promote cassette exon inclusion. This interaction enhances metastatic lung colonization and cancer cell invasion, in part through SNRPA1-mediated regulation of PLEC alternative splicing, which can be counteracted by splicing modulating morpholinos. Our findings establish a noncanonical regulatory role for SNRPA1 as a prometastatic splicing enhancer in breast cancer., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
7. Future avenues for Alzheimer's disease detection and therapy: liquid biopsy, intracellular signaling modulation, systems pharmacology drug discovery.
- Author
-
Hampel H, Vergallo A, Caraci F, Cuello AC, Lemercier P, Vellas B, Giudici KV, Baldacci F, Hänisch B, Haberkamp M, Broich K, Nisticò R, Emanuele E, Llavero F, Zugaza JL, Lucía A, Giacobini E, and Lista S
- Subjects
- Alzheimer Disease metabolism, Animals, Anti-Inflammatory Agents administration & dosage, Anti-Inflammatory Agents metabolism, Drug Discovery methods, Drug Repositioning methods, Drug Repositioning trends, Forecasting, Humans, Intracellular Fluid metabolism, Liquid Biopsy methods, Liquid Biopsy trends, Membrane Glycoproteins metabolism, Pharmacology, Clinical methods, Receptors, Immunologic metabolism, Signal Transduction drug effects, Signal Transduction physiology, Systems Biology methods, Alzheimer Disease diagnosis, Alzheimer Disease drug therapy, Drug Discovery trends, Intracellular Fluid drug effects, Pharmacology, Clinical trends, Systems Biology trends
- Abstract
When Alzheimer's disease (AD) disease-modifying therapies will be available, global healthcare systems will be challenged by a large-scale demand for clinical and biological screening. Validation and qualification of globally accessible, minimally-invasive, and time-, cost-saving blood-based biomarkers need to be advanced. Novel pathophysiological mechanisms (and related candidate biomarkers) - including neuroinflammation pathways (TREM2 and YKL-40), axonal degeneration (neurofilament light chain protein), synaptic dysfunction (neurogranin, synaptotagmin, α-synuclein, and SNAP-25) - may be integrated into an expanding pathophysiological and biomarker matrix and, ultimately, integrated into a comprehensive blood-based liquid biopsy, aligned with the evolving ATN + classification system and the precision medicine paradigm. Liquid biopsy-based diagnostic and therapeutic algorithms are increasingly employed in Oncology disease-modifying therapies and medical practice, showing an enormous potential for AD and other brain diseases as well. For AD and other neurodegenerative diseases, newly identified aberrant molecular pathways have been identified as suitable therapeutic targets and are currently investigated by academia/industry-led R&D programs, including the nerve-growth factor pathway in basal forebrain cholinergic neurons, the sigma1 receptor, and the GTPases of the Rho family. Evidence for a clinical long-term effect on cognitive function and brain health span of cholinergic compounds, drug candidates for repositioning programs, and non-pharmacological multidomain interventions (nutrition, cognitive training, and physical activity) is developing as well. Ultimately, novel pharmacological paradigms, such as quantitative systems pharmacology-based integrative/explorative approaches, are gaining momentum to optimize drug discovery and accomplish effective pathway-based strategies for precision medicine. This article is part of the special issue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
8. RBMS1 Suppresses Colon Cancer Metastasis through Targeted Stabilization of Its mRNA Regulon.
- Author
-
Yu J, Navickas A, Asgharian H, Culbertson B, Fish L, Garcia K, Olegario JP, Dermit M, Dodel M, Hänisch B, Luo Y, Weinberg EM, Dienstmann R, Warren RS, Mardakheh FK, and Goodarzi H
- Subjects
- Animals, Cell Line, Tumor, Colon pathology, Colonic Neoplasms diagnosis, Colonic Neoplasms genetics, DNA-Binding Proteins genetics, Gene Knockdown Techniques, Humans, Intestinal Mucosa pathology, Liver Neoplasms secondary, Male, Mice, Neoplasm Staging, RNA Stability genetics, RNA-Binding Proteins genetics, RNA-Seq, Regulon, Xenograft Model Antitumor Assays, Colonic Neoplasms pathology, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Liver Neoplasms genetics, RNA, Messenger genetics, RNA-Binding Proteins metabolism
- Abstract
Identifying master regulators that drive pathologic gene expression is a key challenge in precision oncology. Here, we have developed an analytic framework, named PRADA, that identifies oncogenic RNA-binding proteins through the systematic detection of coordinated changes in their target regulons. Application of this approach to data collected from clinical samples, patient-derived xenografts, and cell line models of colon cancer metastasis revealed the RNA-binding protein RBMS1 as a suppressor of colon cancer progression. We observed that silencing RBMS1 results in increased metastatic capacity in xenograft mouse models, and that restoring its expression blunts metastatic liver colonization. We have found that RBMS1 functions as a posttranscriptional regulator of RNA stability by directly binding its target mRNAs. Together, our findings establish a role for RBMS1 as a previously unknown regulator of RNA stability and as a suppressor of colon cancer metastasis with clinical utility for risk stratification of patients. SIGNIFICANCE: By applying a new analytic approach to transcriptomic data from clinical samples and models of colon cancer progression, we have identified RBMS1 as a suppressor of metastasis and as a post-transcriptional regulator of RNA stability. Notably, RBMS1 silencing and downregulation of its targets are negatively associated with patient survival. See related commentary by Carter, p. 1261 . This article is highlighted in the In This Issue feature, p. 1241 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
9. DEGS1-associated aberrant sphingolipid metabolism impairs nervous system function in humans.
- Author
-
Karsai G, Kraft F, Haag N, Korenke GC, Hänisch B, Othman A, Suriyanarayanan S, Steiner R, Knopp C, Mull M, Bergmann M, Schröder JM, Weis J, Elbracht M, Begemann M, Hornemann T, and Kurth I
- Subjects
- Amino Acid Substitution, Cell Line, Female, Humans, Male, Exome Sequencing, Central Nervous System Diseases enzymology, Central Nervous System Diseases genetics, Central Nervous System Diseases pathology, Fatty Acid Desaturases genetics, Fatty Acid Desaturases metabolism, Lipid Metabolism, Inborn Errors enzymology, Lipid Metabolism, Inborn Errors genetics, Lipid Metabolism, Inborn Errors pathology, Mutation, Missense, Sphingosine genetics, Sphingosine metabolism
- Abstract
Background: Sphingolipids are important components of cellular membranes and functionally associated with fundamental processes such as cell differentiation, neuronal signaling, and myelin sheath formation. Defects in the synthesis or degradation of sphingolipids leads to various neurological pathologies; however, the entire spectrum of sphingolipid metabolism disorders remains elusive., Methods: A combined approach of genomics and lipidomics was applied to identify and characterize a human sphingolipid metabolism disorder., Results: By whole-exome sequencing in a patient with a multisystem neurological disorder of both the central and peripheral nervous systems, we identified a homozygous p.Ala280Val variant in DEGS1, which catalyzes the last step in the ceramide synthesis pathway. The blood sphingolipid profile in the patient showed a significant increase in dihydro sphingolipid species that was further recapitulated in patient-derived fibroblasts, in CRISPR/Cas9-derived DEGS1-knockout cells, and by pharmacological inhibition of DEGS1. The enzymatic activity in patient fibroblasts was reduced by 80% compared with wild-type cells, which was in line with a reduced expression of mutant DEGS1 protein. Moreover, an atypical and potentially neurotoxic sphingosine isomer was identified in patient plasma and in cells expressing mutant DEGS1., Conclusion: We report DEGS1 dysfunction as the cause of a sphingolipid disorder with hypomyelination and degeneration of both the central and peripheral nervous systems., Trial Registration: Not applicable., Funding: Seventh Framework Program of the European Commission, Swiss National Foundation, Rare Disease Initiative Zurich.
- Published
- 2019
- Full Text
- View/download PDF
10. Molecular basis for the antiproliferative effect of agmatine in tumor cells of colonic, hepatic, and neuronal origin.
- Author
-
Wolf C, Brüss M, Hänisch B, Göthert M, von Kügelgen I, and Molderings GJ
- Subjects
- Actins genetics, Animals, Base Sequence, Brain Neoplasms, Carcinoma, Hepatocellular, Cell Line, Tumor, Colonic Neoplasms, DNA Primers, Enzyme Inhibitors pharmacology, Humans, Liver Neoplasms, Ornithine Decarboxylase genetics, Rats, Agmatine pharmacology, Cell Division drug effects
- Abstract
The aim of the present study was to challenge potential mechanisms of action underlying the inhibition of tumor cell proliferation by agmatine. Agmatine inhibited proliferation of the human hepatoma cells HepG2, the human adenocarcinoma cells HT29, the rat hepatoma cells McRH7777, and the rat pheochromocytoma cells PC-12. Inhibition of proliferation of HepG2 cells was associated with an abolition of expression of ornithine decarboxylase (ODC) protein and a doubling of mRNA content encoding ODC. In HepG2 cells, silencing of ODC-antizyme-1, but not of antizyme inhibitor, by RNA interference resulted in an increase of agmatine's antiproliferative effect. Thus, the distinct decrease in intracellular polyamine content by agmatine was due to a reduced translation of the synthesizing protein ODC but was not essentially mediated by induction of ODC-antizyme or blockade of antizyme inhibitor. In interaction experiments 1 mM L-arginine, 1 mM D-arginine, 1 mM citrulline, 100 microM N(omega)-nitro-L-arginine methyl ester, 1 and 10 microM sodium nitroprusside, and 1 microM N1-guanyl-1,7-diaminoheptane failed to alter agmatine's antiproliferative effect. Hence, the antiproliferative effect of agmatine in HT29 and HepG2 cells is due to an interaction with neither the NO synthases, the hypusination of eIF5A, nor an agmatine-induced reduction in availability of intracellular L-arginine. L-Arginine and citrulline, but not d-arginine, inhibited tumor cell proliferation by themselves. Their inhibitory effect was abolished after silencing of arginine decarboxylase (ADC) expression by RNA interference indicating the conversion to agmatine by ADC. Finally, in the four cell lines under study, agmatine-induced inhibition of cell proliferation was paralleled by an increase in intracellular caspase-3 activity, indicating a promotion of apoptosis.
- Published
- 2007
- Full Text
- View/download PDF
11. Fulminant meningococcal meningitis and sepsis associated with severe hypothyroidism caused by autoimmune (Hashimoto) thyreoiditis.
- Author
-
Mayer D, Miller-Hänisch B, Böckh A, and Hehrmann R
- Subjects
- Adult, Bacteremia immunology, Germany, Humans, Hypothyroidism immunology, Male, Meningitis, Meningococcal immunology, Sri Lanka ethnology, Bacteremia complications, Hypothyroidism complications, Meningitis, Meningococcal complications, Thyroiditis, Autoimmune complications
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.