Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Aging Dis ; 15(1): 226-244, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37962464

ABSTRACT

Diets that restrict caloric or protein intake offer a variety of benefits, including decreasing the incidence of cancer. However, whether such diets pose a substantial therapeutic benefit as auxiliary cancer treatments remains unclear. We determined the effects of severe protein depletion on tumorigenesis in a Drosophila melanogaster intestinal tumor model, using a human RAF gain-of-function allele. Severe and continuous protein restriction significantly reduced tumor growth but resulted in premature death. Therefore, we developed a diet in which short periods of severe protein restriction alternated cyclically with periods of complete feeding. This nutritional regime reduced tumor mass, restored gut functionality, and rescued the lifespan of oncogene-expressing flies to the levels observed in healthy flies on a continuous, fully nutritious diet. Furthermore, this diet reduced the chemotherapy-induced stem cell activity associated with tumor recurrence. Transcriptome analysis revealed long-lasting changes in the expression of key genes involved in multiple major developmental signaling pathways. Overall, the data suggest that recurrent severe protein depletion effectively mimics the health benefits of continuous protein restriction, without undesired nutritional shortcomings. This provides seminal insights into the mechanisms of the memory effect required to maintain the positive effects of protein restriction throughout the phases of a full diet. Finally, the repetitive form of strict protein restriction is an ideal strategy for adjuvant cancer therapy that is useful in many tumor contexts.


Subject(s)
Drosophila , Intestinal Neoplasms , Animals , Humans , Longevity/genetics , Drosophila melanogaster/genetics , Caloric Restriction , Neoplasm Recurrence, Local , Intestinal Neoplasms/genetics
2.
Front Oncol ; 13: 1200897, 2023.
Article in English | MEDLINE | ID: mdl-37384296

ABSTRACT

Introduction: Resistance in anti-cancer treatment is a result of clonal evolution and clonal selection. In chronic myeloid leukemia (CML), the hematopoietic neoplasm is predominantly caused by the formation of the BCR::ABL1 kinase. Evidently, treatment with tyrosine kinase inhibitors (TKIs) is tremendously successful. It has become the role model of targeted therapy. However, therapy resistance to TKIs leads to loss of molecular remission in about 25% of CML patients being partially due to BCR::ABL1 kinase mutations, while for the remaining cases, various other mechanisms are discussed. Methods: Here, we established an in vitro-TKI resistance model against the TKIs imatinib and nilotinib and performed exome sequencing. Results: In this model, acquired sequence variants in NRAS, KRAS, PTPN11, and PDGFRB were identified in TKI resistance. The well-known pathogenic NRAS p.(Gln61Lys) variant provided a strong benefit for CML cells under TKI exposure visible by increased cell number (6.2-fold, p < 0.001) and decreased apoptosis (-25%, p < 0.001), proving the functionality of our approach. The transfection of PTPN11 p.(Tyr279Cys) led to increased cell number (1.7-fold, p = 0.03) and proliferation (2.0-fold, p < 0.001) under imatinib treatment. Discussion: Our data demonstrate that our in vitro-model can be used to study the effect of specific variants on TKI resistance and to identify new driver mutations and genes playing a role in TKI resistance. The established pipeline can be used to study candidates acquired in TKI-resistant patients, thereby providing new options for the development of new therapy strategies to overcome resistance.

3.
Cells ; 12(11)2023 05 27.
Article in English | MEDLINE | ID: mdl-37296610

ABSTRACT

The aggressive features of glioblastoma (GBM) are associated with dormancy. Our previous transcriptome analysis revealed that several genes were regulated during temozolomide (TMZ)-promoted dormancy in GBM. Focusing on genes involved in cancer progression, Chemokine (C-C motif) Receptor-Like (CCRL)1, Schlafen (SLFN)13, Sloan-Kettering Institute (SKI), Cdk5 and Abl Enzyme Substrate (Cables)1, and Dachsous Cadherin-Related (DCHS)1 were selected for further validation. All showed clear expression and individual regulatory patterns under TMZ-promoted dormancy in human GBM cell lines, patient-derived primary cultures, glioma stem-like cells (GSCs), and human GBM ex vivo samples. All genes exhibited complex co-staining patterns with different stemness markers and with each other, as examined by immunofluorescence staining and underscored by correlation analyses. Neurosphere formation assays revealed higher numbers of spheres during TMZ treatment, and gene set enrichment analysis of transcriptome data revealed significant regulation of several GO terms, including stemness-associated ones, indicating an association between stemness and dormancy with the involvement of SKI. Consistently, inhibition of SKI during TMZ treatment resulted in higher cytotoxicity, proliferation inhibition, and lower neurosphere formation capacity compared to TMZ alone. Overall, our study suggests the involvement of CCRL1, SLFN13, SKI, Cables1, and DCHS1 in TMZ-promoted dormancy and demonstrates their link to stemness, with SKI being particularly important.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Temozolomide/pharmacology , Glioblastoma/drug therapy , Glioblastoma/genetics , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic
4.
J Autoimmun ; 135: 102985, 2023 02.
Article in English | MEDLINE | ID: mdl-36621173

ABSTRACT

Autoimmune Encephalitis (AE) spans a group of non-infectious inflammatory conditions of the central nervous system due to an imbalanced immune response. Aiming to elucidate the pathophysiological mechanisms of AE, we applied an unsupervised proteomic approach to analyze the cerebrospinal fluid (CSF) protein profile of AE patients with autoantibodies against N-methyl-d-aspartate receptor (NMDAR) (n = 9), leucine-rich glioma-inactivated protein 1 (LGI1) (n = 9), or glutamate decarboxylase 65 (GAD65) (n = 8) compared to 9 patients with relapsing-remitting multiple sclerosis as inflammatory controls, and 10 patients with somatic symptom disorder as non-inflammatory controls. We found a dysregulation of the complement system, a disbalance between pro-inflammatory and anti-inflammatory proteins on the one hand, and dysregulation of proteins involved in synaptic transmission, synaptogenesis, brain connectivity, and neurodegeneration on the other hand to a different extent in all AE subtypes compared to non-inflammatory controls. Furthermore, elevated levels of several proteases and reduction in protease inhibitors could be detected in all AE subtypes compared to non-inflammatory controls. Moreover, the different AE subtypes showed distinct protein profiles compared to each other and inflammatory controls which may facilitate future identification of disease-specific biomarkers. Overall, CSF proteomics provides insights into the complex pathophysiological mechanisms of AE, including immune dysregulation, neuronal dysfunction, neurodegeneration, and altered protease function.


Subject(s)
Encephalitis , Multiple Sclerosis, Relapsing-Remitting , Humans , Proteomics , Proteins , Autoantibodies
5.
Sci Rep ; 12(1): 3728, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260584

ABSTRACT

To elucidate cross-sectional patterns and longitudinal changes of oral and stool microbiota in multiple sclerosis (MS) patients and the effect of B-cell depletion. We conducted an observational, longitudinal clinical cohort study analysing four timepoints over 12 months in 36 MS patients, of whom 22 initiated B-cell depleting therapy with ocrelizumab and a healthy control group. For microbiota analysis of the oral cavity and the gut, provided stool and oral swab samples underwent 16S rDNA sequencing and subsequent bioinformatic analyses. Oral microbiota-patterns exhibited a reduced alpha-diversity and unique differential microbiota changes compared to stool such as increased levels of Proteobacteria and decreased abundance of Actinobacteria. Following B-cell depletion, we observed increased alpha-diversity in the gut and the oral cavity as well as a long-term sustained reduction of pro-inflammatory Gram-negative bacteria (e.g., Escherichia/Shigella). MS patients have altered stool and oral microbiota diversity patterns compared to healthy controls, which are most pronounced in patients with higher disease activity and disability. Therapeutic B-cell depletion is associated with persisting regression of these changes. Whether these microbial changes are unspecific side-effects of B-cell depletion or indirectly modulate MS disease activity and progression is currently unknown and necessitates further investigations.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Multiple Sclerosis , Cohort Studies , Cross-Sectional Studies , Dysbiosis/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Humans , Multiple Sclerosis/drug therapy , RNA, Ribosomal, 16S/genetics
6.
Immunity ; 53(6): 1258-1271.e5, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33296686

ABSTRACT

CD4+ T cells reactive against SARS-CoV-2 can be found in unexposed individuals, and these are suggested to arise in response to common cold coronavirus (CCCoV) infection. Here, we utilized SARS-CoV-2-reactive CD4+ T cell enrichment to examine the antigen avidity and clonality of these cells, as well as the relative contribution of CCCoV cross-reactivity. SARS-CoV-2-reactive CD4+ memory T cells were present in virtually all unexposed individuals examined, displaying low functional avidity and multiple, highly variable cross-reactivities that were not restricted to CCCoVs. SARS-CoV-2-reactive CD4+ T cells from COVID-19 patients lacked cross-reactivity to CCCoVs, irrespective of strong memory T cell responses against CCCoV in all donors analyzed. In severe but not mild COVID-19, SARS-CoV-2-specific T cells displayed low functional avidity and clonality, despite increased frequencies. Our findings identify low-avidity CD4+ T cell responses as a hallmark of severe COVID-19 and argue against a protective role for CCCoV-reactive T cells in SARS-CoV-2 infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , COVID-19/immunology , Receptors, Antigen, T-Cell/metabolism , Rhinovirus/immunology , SARS-CoV-2/immunology , Antigens, Viral/immunology , Cells, Cultured , Cross Reactions , Disease Progression , Environmental Exposure , Humans , Immunologic Memory , Lymphocyte Activation , Protein Binding , Severity of Illness Index , T-Cell Antigen Receptor Specificity
7.
Cell ; 178(6): 1299-1312.e29, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474368

ABSTRACT

Metformin is the first-line therapy for treating type 2 diabetes and a promising anti-aging drug. We set out to address the fundamental question of how gut microbes and nutrition, key regulators of host physiology, affect the effects of metformin. Combining two tractable genetic models, the bacterium E. coli and the nematode C. elegans, we developed a high-throughput four-way screen to define the underlying host-microbe-drug-nutrient interactions. We show that microbes integrate cues from metformin and the diet through the phosphotransferase signaling pathway that converges on the transcriptional regulator Crp. A detailed experimental characterization of metformin effects downstream of Crp in combination with metabolic modeling of the microbiota in metformin-treated type 2 diabetic patients predicts the production of microbial agmatine, a regulator of metformin effects on host lipid metabolism and lifespan. Our high-throughput screening platform paves the way for identifying exploitable drug-nutrient-microbiome interactions to improve host health and longevity through targeted microbiome therapies. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Microbiome/drug effects , Host Microbial Interactions/drug effects , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Agmatine/metabolism , Animals , Caenorhabditis elegans/microbiology , Cyclic AMP Receptor Protein , Escherichia coli/drug effects , Escherichia coli/genetics , Humans , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Longevity/drug effects , Metformin/pharmacology , Nutrients/metabolism
8.
Nat Commun ; 10(1): 2459, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31150008

ABSTRACT

The original version of this Article contained an error in the spelling of the author Jule Müller, which was incorrectly given as Julia Müller. Additionally, in Fig. 4a, the blue-red colour scale for fold change in ageing/disease regulation included a blue stripe in place of a red stripe at the right-hand end of the scale. These errors have been corrected in both the PDF and HTML versions of the Article.

9.
Cancer Lett ; 453: 95-106, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30930235

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is commonly diagnosed when liver metastases already emerged. We recently demonstrated that hepatic stromal cells determine the dormancy status along with cancer stem cell (CSC) properties of pancreatic ductal epithelial cells (PDECs) during metastasis. This study investigated the influence of the hepatic microenvironment - and its inflammatory status - on metabolic alterations and how these impact cell growth and CSC-characteristics of PDECs. Coculture with hepatic stellate cells (HSCs), simulating a physiological liver stroma, but not with hepatic myofibroblasts (HMFs) representing liver inflammation promoted expression of Succinate Dehydrogenase subunit B (SDHB) and an oxidative metabolism along with a quiescent phenotype in PDECs. SiRNA-mediated SDHB knockdown increased cell growth and CSC-properties. Moreover, liver micrometastases of tumor bearing KPC mice strongly expressed SDHB while expression of the CSC-marker Nestin was exclusively found in macrometastases. Consistently, RNA-sequencing and in silico modeling revealed significantly altered metabolic fluxes and enhanced SDH activity predominantly in premalignant PDECs in the presence of HSC compared to HMF. Overall, these data emphasize that the hepatic microenvironment determines the metabolism of disseminated PDECs thereby controlling cell growth and CSC-properties during liver metastasis.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Coculture Techniques , Down-Regulation , Humans , Mice , Neoplasm Metastasis , Neoplasm Micrometastasis , Neoplastic Stem Cells/metabolism , Oxidative Phosphorylation , Stromal Cells/metabolism , Stromal Cells/pathology , Succinate Dehydrogenase/metabolism
10.
Gastroenterology ; 156(1): 145-159.e19, 2019 01.
Article in English | MEDLINE | ID: mdl-30273559

ABSTRACT

BACKGROUND & AIMS: RNase H2 is a holoenzyme, composed of 3 subunits (ribonuclease H2 subunits A, B, and C), that cleaves RNA:DNA hybrids and removes mis-incorporated ribonucleotides from genomic DNA through ribonucleotide excision repair. Ribonucleotide incorporation by eukaryotic DNA polymerases occurs during every round of genome duplication and produces the most frequent type of naturally occurring DNA lesion. We investigated whether intestinal epithelial proliferation requires RNase H2 function and whether RNase H2 activity is disrupted during intestinal carcinogenesis. METHODS: We generated mice with epithelial-specific deletion of ribonuclease H2 subunit B (H2bΔIEC) and mice that also had deletion of tumor-suppressor protein p53 (H2b/p53ΔIEC); we compared phenotypes with those of littermate H2bfl/fl or H2b/p53fl/fl (control) mice at young and old ages. Intestinal tissues were collected and analyzed by histology. We isolated epithelial cells, generated intestinal organoids, and performed RNA sequence analyses. Mutation signatures of spontaneous tumors from H2b/p53ΔIEC mice were characterized by exome sequencing. We collected colorectal tumor specimens from 467 patients, measured levels of ribonuclease H2 subunit B, and associated these with patient survival times and transcriptome data. RESULTS: The H2bΔIEC mice had DNA damage to intestinal epithelial cells and proliferative exhaustion of the intestinal stem cell compartment compared with controls and H2b/p53ΔIEC mice. However, H2b/p53ΔIEC mice spontaneously developed small intestine and colon carcinomas. DNA from these tumors contained T>G base substitutions at GTG trinucleotides. Analyses of transcriptomes of human colorectal tumors associated lower levels of RNase H2 with shorter survival times. CONCLUSIONS: In analyses of mice with disruption of the ribonuclease H2 subunit B gene and colorectal tumors from patients, we provide evidence that RNase H2 functions as a colorectal tumor suppressor. H2b/p53ΔIEC mice can be used to study the roles of RNase H2 in tissue-specific carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/enzymology , Genomic Instability , Intestinal Neoplasms/prevention & control , Intestine, Small/enzymology , Ribonuclease H/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/chemically induced , Colitis/enzymology , Colitis/genetics , Colitis/pathology , DNA Damage , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/pathology , Female , Genetic Predisposition to Disease , Humans , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Mice, Knockout , Phenotype , Ribonuclease H/deficiency , Ribonuclease H/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
11.
J Innate Immun ; 11(5): 393-404, 2019.
Article in English | MEDLINE | ID: mdl-30566939

ABSTRACT

Animals are usually regarded as independent entities within their respective environments. However, within an organism, eukaryotes and prokaryotes interact dynamically to form the so-called metaorganism or holobiont, where each partner fulfils its versatile and crucial role. This review focuses on the interplay between microorganisms and multicellular eukaryotes in the context of host physiology, in particular aging and mucus-associated crosstalk. In addition to the interactions between bacteria and the host, we highlight the importance of viruses and nonmodel organisms. Moreover, we discuss current culturing and computational methodologies that allow a deeper understanding of underlying mechanisms controlling the physiology of metaorganisms.


Subject(s)
Host Microbial Interactions/physiology , Microbiota/physiology , Aging , Animals , Computational Biology , Health Status , Humans , Models, Biological , Mucus/microbiology , Mucus/virology , Symbiosis/physiology
12.
Nat Commun ; 9(1): 327, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382830

ABSTRACT

Disease epidemiology during ageing shows a transition from cancer to degenerative chronic disorders as dominant contributors to mortality in the old. Nevertheless, it has remained unclear to what extent molecular signatures of ageing reflect this phenomenon. Here we report on the identification of a conserved transcriptomic signature of ageing based on gene expression data from four vertebrate species across four tissues. We find that ageing-associated transcriptomic changes follow trajectories similar to the transcriptional alterations observed in degenerative ageing diseases but are in opposite direction to the transcriptomic alterations observed in cancer. We confirm the existence of a similar antagonism on the genomic level, where a majority of shared risk alleles which increase the risk of cancer decrease the risk of chronic degenerative disorders and vice versa. These results reveal a fundamental trade-off between cancer and degenerative ageing diseases that sheds light on the pronounced shift in their epidemiology during ageing.


Subject(s)
Aging/genetics , Cardiovascular Diseases/genetics , Diabetes Mellitus/genetics , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Transcriptome , Adolescent , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Animals , Brain/growth & development , Brain/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Child , Child, Preschool , Chronic Disease , Diabetes Mellitus/blood , Diabetes Mellitus/pathology , Fundulidae/genetics , Fundulidae/growth & development , Fundulidae/metabolism , Gene Ontology , Genome, Human , Humans , Infant , Liver/growth & development , Liver/metabolism , Mice , Middle Aged , Molecular Sequence Annotation , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/pathology , Skin/growth & development , Skin/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
13.
BMC Genomics ; 18(1): 517, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28683819

ABSTRACT

BACKGROUND: Gastric cancer is the fourth most common cancer and the second leading cause of cancer death worldwide. In order to understand the genetic background, we sequenced the whole exome and the whole genome of one microsatellite stable as well as one microsatellite unstable tumor and the matched healthy tissue on two different NGS platforms. We here aimed to provide a comparative approach for individual clinical tumor sequencing and annotation using different sequencing technologies and mutation calling algorithms. RESULTS: We applied a population-based whole genome resource as a novel pathway-based filter for interpretation of genomic alterations from single nucleotide variations (SNV), indels, and large structural variations. In addition to a comparison with tumor genome database resources and a filtering approach using data from the 1000 Genomes Project, we performed pyrosequencing analysis and immunohistochemistry in a large cohort of 428 independent gastric cancer cases. CONCLUSION: We here provide an example comparing the usefulness and potential pitfalls of different technologies for a clinical interpretation of genomic sequence data of individual gastric cancer samples. Using different filtering approaches, we identified a multitude of novel potentially damaging mutations and could show a validated association between a mutation in GNAS and gastric cancer.


Subject(s)
Exome Sequencing , Stomach Neoplasms/genetics , Aged , Chromogranins/genetics , Cohort Studies , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation, Neoplastic , Humans , INDEL Mutation , Male , Polymorphism, Single Nucleotide
14.
BMC Genomics ; 18(1): 329, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446171

ABSTRACT

BACKGROUND: The phenomenon of immune priming, i.e. enhanced protection following a secondary exposure to a pathogen, has now been demonstrated in a wide range of invertebrate species. Despite accumulating phenotypic evidence, knowledge of its mechanistic underpinnings is currently very limited. Here we used the system of the red flour beetle, Tribolium castaneum and the insect pathogen Bacillus thuringiensis (Bt) to further our molecular understanding of the oral immune priming phenomenon. We addressed how ingestion of bacterial cues (derived from spore supernatants) of an orally pathogenic and non-pathogenic Bt strain affects gene expression upon later challenge exposure, using a whole-transcriptome sequencing approach. RESULTS: Whereas gene expression of individuals primed with the orally non-pathogenic strain showed minor changes to controls, we found that priming with the pathogenic strain induced regulation of a large set of distinct genes, many of which are known immune candidates. Intriguingly, the immune repertoire activated upon priming and subsequent challenge qualitatively differed from the one mounted upon infection with Bt without previous priming. Moreover, a large subset of priming-specific genes showed an inverse regulation compared to their regulation upon challenge only. CONCLUSIONS: Our data demonstrate that gene expression upon infection is strongly affected by previous immune priming. We hypothesise that this shift in gene expression indicates activation of a more targeted and efficient response towards a previously encountered pathogen, in anticipation of potential secondary encounter.


Subject(s)
Bacillus thuringiensis/physiology , Gene Expression Regulation/immunology , Larva/immunology , Larva/microbiology , Tribolium/immunology , Tribolium/microbiology , Administration, Oral , Animals , Larva/genetics , Species Specificity , Tribolium/genetics
15.
Gut ; 66(12): 2087-2097, 2017 12.
Article in English | MEDLINE | ID: mdl-27694142

ABSTRACT

OBJECTIVE: An inadequate host response to the intestinal microbiota likely contributes to the manifestation and progression of human inflammatory bowel disease (IBD). However, molecular approaches to unravelling the nature of the defective crosstalk and its consequences for intestinal metabolic and immunological networks are lacking. We assessed the mucosal transcript levels, splicing architecture and mucosa-attached microbial communities of patients with IBD to obtain a comprehensive view of the underlying, hitherto poorly characterised interactions, and how these are altered in IBD. DESIGN: Mucosal biopsies from Crohn's disease and patients with UC, disease controls and healthy individuals (n=63) were subjected to microbiome, transcriptome and splicing analysis, employing next-generation sequencing. The three data levels were integrated by different bioinformatic approaches, including systems biology-inspired network and pathway analysis. RESULTS: Microbiota, host transcript levels and host splicing patterns were influenced most strongly by tissue differences, followed by the effect of inflammation. Both factors point towards a substantial disease-related alteration of metabolic processes. We also observed a strong enrichment of splicing events in inflamed tissues, accompanied by an alteration of the mucosa-attached bacterial taxa. Finally, we noted a striking uncoupling of the three molecular entities when moving from healthy individuals via disease controls to patients with IBD. CONCLUSIONS: Our results provide strong evidence that the interplay between microbiome and host transcriptome, which normally characterises a state of intestinal homeostasis, is drastically perturbed in Crohn's disease and UC. Consequently, integrating multiple OMICs levels appears to be a promising approach to further disentangle the complexity of IBD.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , RNA Splicing , Biopsy , Case-Control Studies , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Inflammatory Bowel Diseases/immunology , Male , RNA Splicing/genetics , RNA Splicing/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Transcriptome/genetics , Transcriptome/immunology
16.
Dev Comp Immunol ; 51(1): 1-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25720978

ABSTRACT

Pathogen infection can activate multiple signaling cascades that ultimately alter the abundance of molecules in cells. This change can be measured both at the transcript and protein level. Studies analyzing the immune response at both levels are, however, rare. Here, we compare transcriptome and proteome data generated after infection of the nematode and model organism Caenorhabditis elegans with the Gram-positive pathogen Bacillus thuringiensis. Our analysis revealed a high overlap between abundance changes of corresponding transcripts and gene products, especially for genes encoding C-type lectin domain-containing proteins, indicating their particular role in worm immunity. We additionally identified a unique signature at the proteome level, suggesting that the C. elegans response to infection is shaped by changes beyond transcription. Such effects appear to be influenced by AMP-activated protein kinases (AMPKs), which may thus represent previously unknown regulators of C. elegans immune defense.


Subject(s)
Bacillus thuringiensis/immunology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans , Gram-Positive Bacterial Infections/immunology , Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Immunity, Innate/genetics , Lectins, C-Type/genetics , Proteome , RNA Processing, Post-Transcriptional , Species Specificity , Transcriptome
17.
BMC Genomics ; 15: 445, 2014 Jun 07.
Article in English | MEDLINE | ID: mdl-24908078

ABSTRACT

BACKGROUND: Pathogens can infect their hosts through different routes. For studying the consequences for host resistance, we here used the entomopathogen Bacillus thuringiensis and the red flour beetle Tribolium castaneum for oral and systemic (i. e. pricking the cuticle) experimental infection. In order to characterize the molecular mechanisms underpinning the two different infection routes, the transcriptomes of beetles of two different T. castaneum populations--one recently collected population (Cro1) and a commonly used laboratory strain (SB)--were analyzed using a next generation RNA sequencing approach. RESULTS: The genetically more diverse population Cro1 showed a significantly larger number of differentially expressed genes. While both populations exhibited similar reactions to pricking, their expression patterns in response to oral infection differed remarkably. In particular, the Cro1 population showed a strong response of cuticular proteins and developmental genes, which might indicate an adaptive developmental flexibility that was lost in the SB population presumably as a result of inbreeding. The immune response of SB was primarily based on antimicrobial peptides, while Cro1 relied on responses mediated by phenoloxidase and reactive oxygen species, which may explain the higher resistance of this strain against oral infection. CONCLUSIONS: Our data demonstrate that immunological and physiological processes underpinning the two different routes of infection are clearly distinct, and that host populations particularly differ in responses to oral infection. Furthermore, gene expression upon pricking infection entailed a strong signal of wounding, highlighting the importance of pricking controls in future infection studies.


Subject(s)
Bacillus thuringiensis/pathogenicity , Coleoptera/genetics , Coleoptera/microbiology , Host-Pathogen Interactions , Animals , Cluster Analysis , Coleoptera/immunology , Coleoptera/metabolism , Computational Biology , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Signal Transduction , Time Factors , Transcriptome
18.
J Exp Med ; 210(10): 2041-56, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24043762

ABSTRACT

Unresolved endoplasmic reticulum (ER) stress in the epithelium can provoke intestinal inflammation. Hypomorphic variants of ER stress response mediators, such as X-box-binding protein 1 (XBP1), confer genetic risk for inflammatory bowel disease. We report here that hypomorphic Xbp1 function instructs a multilayered regenerative response in the intestinal epithelium. This is characterized by intestinal stem cell (ISC) expansion as shown by an inositol-requiring enzyme 1α (Ire1α)-mediated increase in Lgr5(+) and Olfm4(+) ISCs and a Stat3-dependent increase in the proliferative output of transit-amplifying cells. These consequences of hypomorphic Xbp1 function are associated with an increased propensity to develop colitis-associated and spontaneous adenomatous polyposis coli (APC)-related tumors of the intestinal epithelium, which in the latter case is shown to be dependent on Ire1α. This study reveals an unexpected role for Xbp1 in suppressing tumor formation through restraint of a pathway that involves an Ire1α- and Stat3-mediated regenerative response of the epithelium as a consequence of ER stress. As such, Xbp1 in the intestinal epithelium not only regulates local inflammation but at the same time also determines the propensity of the epithelium to develop tumors.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Stress , Intestinal Mucosa/metabolism , Intestines/pathology , Stem Cells/metabolism , Stem Cells/pathology , Transcription Factors/genetics , Animals , Autocrine Communication/genetics , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Enzyme Activation , Gene Deletion , Genes, APC , Interleukin-11/metabolism , Interleukin-6/metabolism , Intestinal Mucosa/pathology , Janus Kinase 1/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , Mice , Mice, Knockout , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Regulatory Factor X Transcription Factors , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Tumor Burden/genetics , X-Box Binding Protein 1
19.
Nature ; 501(7468): 506-11, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24037378

ABSTRACT

Genome sequencing projects are discovering millions of genetic variants in humans, and interpretation of their functional effects is essential for understanding the genetic basis of variation in human traits. Here we report sequencing and deep analysis of messenger RNA and microRNA from lymphoblastoid cell lines of 462 individuals from the 1000 Genomes Project--the first uniformly processed high-throughput RNA-sequencing data from multiple human populations with high-quality genome sequences. We discover extremely widespread genetic variation affecting the regulation of most genes, with transcript structure and expression level variation being equally common but genetically largely independent. Our characterization of causal regulatory variation sheds light on the cellular mechanisms of regulatory and loss-of-function variation, and allows us to infer putative causal variants for dozens of disease-associated loci. Altogether, this study provides a deep understanding of the cellular mechanisms of transcriptome variation and of the landscape of functional variants in the human genome.


Subject(s)
Genetic Variation/genetics , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Sequence Analysis, RNA , Transcriptome/genetics , Alleles , Cell Line, Transformed , Exons/genetics , Gene Expression Profiling , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics
20.
Bioinformatics ; 29(13): 1600-6, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23620359

ABSTRACT

MOTIVATION: Protocols to generate strand-specific transcriptomes with next-generation sequencing platforms have been used by the scientific community roughly since 2008. Strand-specific reads allow for detection of antisense events and a higher resolution of expression profiles enabling extension of current transcript annotations. However, applications making use of this strandedness information are still scarce. RESULTS: Here we present a tool (Janus), which focuses on the identification of transcriptional active regions in antisense orientation to known and novel transcribed elements of the genome. Janus can compare the antisense events of multiple samples and assigns scores to identify mutual expression of either transcript in a sense/antisense pair, which could hint to regulatory mechanisms. Janus is able to make use of single-nucleotide variant (SNV) and methylation data, if available, and reports the sense to antisense ratio of regions in the vicinity of the identified genetic and epigenetic variation. Janus interrogates positions of heterozygous SNVs to identify strand-specific allelic imbalance. AVAILABILITY: Janus is written in C/C++ and freely available at http://www.ikmb.uni-kiel.de/janus/janus.html under terms of GNU General Public License, for both, Linux and Windows 64×. Although the binaries will work without additional downloads, the software depends on bamtools (https://github.com/pezmaster31/bamtools) for compilation. A detailed tutorial section is included in the first section of the supplemental material and included as brief readme.txt in the tutorial archive. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Expression Profiling , High-Throughput Nucleotide Sequencing , RNA, Antisense/biosynthesis , Software , Cell Line, Transformed , DNA Methylation , Genetic Variation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...