Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 18666, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37907668

ABSTRACT

Several preclinical models have been recently developed for metabolic associated fatty liver disease (MAFLD) and associated hepatocellular carcinoma (HCC) but comprehensive analysis of the regulatory and transcriptional landscapes underlying disease in these models are still missing. We investigated the regulatory and transcriptional landscape in fatty livers and liver tumours from DIAMOND mice that faithfully mimic human HCC development in the context of MAFLD. RNA-sequencing and ChIP-sequencing revealed rewiring of the Wnt/ß-catenin regulatory network in DIAMOND tumours, as manifested by chromatin remodelling and associated switching in the expression of the canonical TCF/LEF downstream effectors. We identified splicing as a major mechanism leading to constitutive oncogenic activation of ß-catenin in a large subset of DIAMOND tumours, a mechanism that is independent on somatic mutations in the locus and that has not been previously shown. Similar splicing events were found in a fraction of human HCC and hepatoblastoma samples.


Subject(s)
Carcinoma, Hepatocellular , Fatty Liver , Liver Neoplasms , Mice , Humans , Animals , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Gene Regulatory Networks , Fatty Liver/genetics , Diet , Wnt Signaling Pathway/genetics
2.
BMC Cancer ; 23(1): 945, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37803333

ABSTRACT

BACKGROUND: Glioblastoma (GB) is the most aggressive of all primary brain tumours and due to its highly invasive nature, surgical resection is nearly impossible. Patients typically rely on radiotherapy with concurrent temozolomide (TMZ) treatment and face a median survival of ~ 14 months. Alterations in the Epidermal Growth Factor Receptor gene (EGFR) are common in GB tumours, but therapies targeting EGFR have not shown significant clinical efficacy. METHODS: Here, we investigated the influence of the EGFR regulatory genome on GB cells and identified novel EGFR enhancers located near the GB-associated SNP rs723527. We used CRISPR/Cas9-based approaches to target the EGFR enhancer regions, generating multiple modified GB cell lines, which enabled us to study the functional response to enhancer perturbation. RESULTS: Epigenomic perturbation of the EGFR regulatory region decreases EGFR expression and reduces the proliferative and invasive capacity of glioblastoma cells, which also undergo a metabolic reprogramming in favour of mitochondrial respiration and present increased apoptosis. Moreover, EGFR enhancer-perturbation increases the sensitivity of GB cells to TMZ, the first-choice chemotherapeutic agent to treat glioblastoma. CONCLUSIONS: Our findings demonstrate how epigenomic perturbation of EGFR enhancers can ameliorate the aggressiveness of glioblastoma cells and enhance the efficacy of TMZ treatment. This study demonstrates how CRISPR/Cas9-based perturbation of enhancers can be used to modulate the expression of key cancer genes, which can help improve the effectiveness of existing cancer treatments and potentially the prognosis of difficult-to-treat cancers such as glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Genes, erbB-1 , ErbB Receptors/metabolism , Epigenomics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Regulatory Sequences, Nucleic Acid
3.
Nat Commun ; 14(1): 6446, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37833281

ABSTRACT

Chromatin organization controls transcription by modulating 3D-interactions between enhancers and promoters in the nucleus. Alterations in epigenetic states and 3D-chromatin organization result in gene expression changes contributing to cancer. Here, we map the promoter-enhancer interactome and regulatory landscape of glioblastoma, the most aggressive primary brain tumour. Our data reveals profound rewiring of promoter-enhancer interactions, chromatin accessibility and redistribution of histone marks in glioblastoma. This leads to loss of long-range regulatory interactions and overall activation of promoters, which orchestrate changes in the expression of genes associated to glutamatergic synapses, axon guidance, axonogenesis and chromatin remodelling. SMAD3 and PITX1 emerge as major transcription factors controlling genes related to synapse organization and axon guidance. Inhibition of SMAD3 and neuronal activity stimulation cooperate to promote proliferation of glioblastoma cells in co-culture with glutamatergic neurons, and in mice bearing patient-derived xenografts. Our findings provide mechanistic insight into the regulatory networks that mediate neurogliomal synaptic communication.


Subject(s)
Glioblastoma , Humans , Animals , Mice , Glioblastoma/genetics , Enhancer Elements, Genetic/genetics , Chromatin , Transcription Factors/genetics , Gene Expression
4.
Cells ; 11(9)2022 04 21.
Article in English | MEDLINE | ID: mdl-35563711

ABSTRACT

Genome architecture, epigenetics and enhancer function control the fate and identity of cells. Reprogramming to induced pluripotent stem cells (iPSCs) changes the transcriptional profile and chromatin landscape of the starting somatic cell to that of the pluripotent cell in a stepwise manner. Changes in the regulatory networks are tightly regulated during normal embryonic development to determine cell fate, and similarly need to function in cell fate control during reprogramming. Switching off the somatic program and turning on the pluripotent program involves a dynamic reorganization of the epigenetic landscape, enhancer function, chromatin accessibility and 3D chromatin topology. Within this context, we will review here the current knowledge on the processes that control the establishment and maintenance of pluripotency during somatic cell reprogramming.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Cellular Reprogramming/genetics , Chromatin/genetics , Epigenesis, Genetic , Epigenomics
5.
Sci Rep ; 11(1): 24410, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34949756

ABSTRACT

AMP-activated protein kinase (AMPK) has an important role in cellular energy homeostasis and has emerged as a promising target for treatment of Type 2 Diabetes (T2D) due to its beneficial effects on insulin sensitivity and glucose homeostasis. O304 is a pan-AMPK activator that has been shown to improve glucose homeostasis in both mouse models of diabetes and in human T2D subjects. Here, we describe the genome-wide transcriptional profile and chromatin landscape of pancreatic islets following O304 treatment of mice fed high-fat diet (HFD). O304 largely prevented genome-wide gene expression changes associated with HFD feeding in CBA mice and these changes were associated with remodelling of active and repressive chromatin marks. In particular, the increased expression of the ß-cell stress marker Aldh1a3 in islets from HFD-mice is completely abrogated following O304 treatment, which is accompanied by loss of active chromatin marks in the promoter as well as distant non-coding regions upstream of the Aldh1a3 gene. Moreover, O304 treatment restored dysfunctional glucose homeostasis as well as expression of key markers associated with ß-cell function in mice with already established obesity. Our findings provide preclinical evidence that O304 is a promising therapeutic compound not only for T2D remission but also for restoration of ß-cell function following remission of T2D diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Enzyme Activators/pharmacology , Gene Expression/drug effects , Histone Code/drug effects , Histone Code/genetics , Histones/metabolism , Islets of Langerhans/metabolism , Obesity/metabolism , Thiadiazoles/pharmacology , AMP-Activated Protein Kinases/physiology , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Glucose/metabolism , Homeostasis/drug effects , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred CBA , Obesity/etiology , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism
6.
Wiley Interdiscip Rev Dev Biol ; 5(2): 169-85, 2016.
Article in English | MEDLINE | ID: mdl-26558551

ABSTRACT

During embryonic development, complex transcriptional programs govern the precision of gene expression. Many key developmental genes are regulated via cis-regulatory elements that are located far away in the linear genome. How sequences located hundreds of kilobases away from a promoter can influence its activity has been the subject of numerous speculations, which all underline the importance of the 3D-organization of the genome. The recent advent of chromosome conformation capture techniques has put into focus the subdivision of the genome into topologically associating domains (TADs). TADs may influence regulatory activities on multiple levels. The relative invariance of TAD limits across cell types suggests that they may form fixed structural domains that could facilitate and/or confine long-range regulatory interactions. However, most recent studies suggest that interactions within TADs are more variable and dynamic than initially described. Hence, different models are emerging regarding how TADs shape the complex 3D conformations, and thereafter influence the networks of cis-interactions that govern gene expression during development. For further resources related to this article, please visit the WIREs website.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Transcriptional Activation , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Promoter Regions, Genetic , Protein Structure, Tertiary
7.
Opt Express ; 21(14): 16584-604, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23938510

ABSTRACT

We demonstrate a technique to improve structural data obtained from Optical Projection Tomography (OPT) using Image Fusion (IF) and contrast normalization. This enables the visualization of molecular expression patterns in biological specimens with highly variable contrast values. In the approach, termed IF-OPT, different exposures are fused by assigning weighted contrasts to each. When applied to projection images from mouse organs and digital phantoms our results demonstrate the capability of IF-OPT to reveal high and low signal intensity details in challenging specimens. We further provide measurements to highlight the benefits of the new algorithm in comparison to other similar methods.


Subject(s)
Algorithms , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Molecular Imaging/methods , Pattern Recognition, Automated/methods , Subtraction Technique , Tomography, Optical/methods , Animals , Mice
8.
Phys Biol ; 10(3): 036009, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23629025

ABSTRACT

The islets of Langerhans, responsible for controlling blood glucose levels, are dispersed within the pancreas. A universal power law governing the fractal spatial distribution of islets in two-dimensional pancreatic sections has been reported. However, the fractal geometry in the actual three-dimensional pancreas volume, and the developmental process that gives rise to such a self-similar structure, has not been investigated. Here, we examined the three-dimensional spatial distribution of islets in intact mouse pancreata using optical projection tomography and found a power law with a fractal dimension of 2.1. Furthermore, based on two-dimensional pancreatic sections of human autopsies, we found that the distribution of human islets also follows a universal power law with a fractal dimension of 1.5 in adult pancreata, which agrees with the value previously reported in smaller mammalian pancreas sections. Finally, we developed a self-avoiding growth model for the development of the islet distribution and found that the fractal nature of the spatial islet distribution may be associated with the self-avoidance in the branching process of vascularization in the pancreas.


Subject(s)
Fractals , Islets of Langerhans/growth & development , Models, Anatomic , Models, Biological , Adult , Animals , Female , Humans , Islets of Langerhans/anatomy & histology , Mice , Mice, Inbred C57BL , Tomography, Optical
9.
J Vis Exp ; (71): e50238, 2013 Jan 12.
Article in English | MEDLINE | ID: mdl-23353681

ABSTRACT

By adapting OPT to include the capability of imaging in the near infrared (NIR) spectrum, we here illustrate the possibility to image larger bodies of pancreatic tissue, such as the rat pancreas, and to increase the number of channels (cell types) that may be studied in a single specimen. We further describe the implementation of a number of computational tools that provide: 1/ accurate positioning of a specimen's (in our case the pancreas) centre of mass (COM) at the axis of rotation (AR); 2/ improved algorithms for post-alignment tuning which prevents geometric distortions during the tomographic reconstruction and 3/ a protocol for intensity equalization to increase signal to noise ratios in OPT-based BCM determinations. In addition, we describe a sample holder that minimizes the risk for unintentional movements of the specimen during image acquisition. Together, these protocols enable assessments of BCM distribution and other features, to be performed throughout the volume of intact pancreata or other organs (e.g. in studies of islet transplantation), with a resolution down to the level of individual islets of Langerhans.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Insulin-Secreting Cells/pathology , Spectroscopy, Near-Infrared/methods , Tomography, Optical/methods , Animals , Mice , Rats
10.
PLoS One ; 6(6): e21753, 2011.
Article in English | MEDLINE | ID: mdl-21738788

ABSTRACT

Despite the extensive use of the mouse as a model for studies of pancreas development and disease, the development of the gastric pancreatic lobe has been largely overlooked. In this study we use optical projection tomography to provide a detailed three-dimensional and quantitative description of pancreatic growth dynamics in the mouse. Hereby, we describe the epithelial and mesenchymal events leading to the formation of the gastric lobe of the pancreas. We show that this structure forms by perpendicular growth from the dorsal pancreatic epithelium into a distinct lateral domain of the dorsal pancreatic mesenchyme. Our data support a role for spleen organogenesis in the establishment of this mesenchymal domain and in mice displaying perturbed spleen development, including Dh +/-, Bapx1-/- and Sox11-/-, gastric lobe development is disturbed. We further show that the expression profile of markers for multipotent progenitors is delayed in the gastric lobe as compared to the splenic and duodenal pancreatic lobes. Altogether, this study provides new information regarding the developmental dynamics underlying the formation of the gastric lobe of the pancreas and recognizes lobular heterogeneities regarding the time course of pancreatic cellular differentiation. Collectively, these data are likely to constitute important elements in future interpretations of the developing and/or diseased pancreas.


Subject(s)
Morphogenesis/physiology , Pancreas/embryology , Spleen/embryology , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Pancreas/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Spleen/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Islets ; 3(4): 204-8, 2011.
Article in English | MEDLINE | ID: mdl-21633198

ABSTRACT

Optical projection tomography (OPT) imaging is a powerful tool for three-dimensional imaging of gene and protein distribution patterns in biomedical specimens. We have previously demonstrated the possibility, by this technique, to extract information of the spatial and quantitative distribution of the islets of Langerhans in the intact mouse pancreas. In order to further increase the sensitivity of OPT imaging for this type of assessment, we have developed a protocol implementing a computational statistical approach: contrast limited adaptive histogram equalization (CLAHE). We demonstrate that this protocol significantly increases the sensitivity of OPT imaging for islet detection, helps preserve islet morphology and diminish subjectivity in thresholding for tomographic reconstruction. When applied to studies of the pancreas from healthy C57BL/6 mice, our data reveal that, at least in this strain, the pancreas harbors substantially more islets than has previously been reported. Further, we provide evidence that the gastric, duodenal and splenic lobes of the pancreas display dramatic differences in total and relative islet and ß-cell mass distribution. This includes a 75% higher islet density in the gastric lobe as compared to the splenic lobe and a higher relative volume of insulin producing cells in the duodenal lobe as compared to the other lobes. Altogether, our data show that CLAHE substantially improves OPT based assessments of the islets of Langerhans and that lobular origin must be taken into careful consideration in quantitative and spatial assessments of the pancreas.


Subject(s)
Computational Biology/methods , Insulin-Secreting Cells/cytology , Islets of Langerhans/anatomy & histology , Tomography, Optical/methods , Animals , Cell Size , Female , Image Enhancement/methods , Imaging, Three-Dimensional/methods , Mice , Mice, Inbred C57BL , Organ Size , Pancreas/anatomy & histology , Pancreas, Exocrine/anatomy & histology , Reproducibility of Results
12.
Dev Biol ; 352(2): 267-77, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21281624

ABSTRACT

Endoderm development is dependent on inductive signals from different structures in close vicinity, including the notochord, lateral plate mesoderm and endothelial cells. Recently, we demonstrated that a functional vascular system is necessary for proper pancreas development, and that sphingosine-1-phosphate (S1P) exhibits the traits of a blood vessel-derived molecule involved in early pancreas morphogenesis. To examine whether S1P(1)-signaling plays a more general role in endoderm development, S1P(1)-deficient mice were analyzed. S1P(1) ablation results in compromised growth of several foregut-derived organs, including the stomach, dorsal and ventral pancreas and liver. Within the developing pancreas the reduction in organ size was due to deficient proliferation of Pdx1(+) pancreatic progenitors, whereas endocrine cell differentiation was unaffected. Ablation of endothelial cells in vitro did not mimic the S1P(1) phenotype, instead, increased organ size and hyperbranching were observed. Consistent with a negative role for endothelial cells in endoderm organ expansion, excessive vasculature was discovered in S1P(1)-deficient embryos. Altogether, our results show that endothelial cell hyperplasia negatively influences organ development in several foregut-derived organs.


Subject(s)
Endoderm/embryology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Embryonic Development , Endoderm/cytology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/genetics , Morphogenesis/physiology , Pancreas/blood supply , Pancreas/cytology , Pancreas/embryology , Pancreas/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors , Trans-Activators/metabolism
13.
Diabetes ; 59(7): 1756-64, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20393145

ABSTRACT

OBJECTIVE: The aim of this study was to refine the information regarding the quantitative and spatial dynamics of infiltrating lymphocytes and remaining beta-cell volume during the progression of type 1 diabetes in the nonobese diabetic (NOD) mouse model of the disease. RESEARCH DESIGN AND METHODS: Using an ex vivo technique, optical projection tomography (OPT), we quantified and assessed the three-dimensional spatial development and progression of insulitis and beta-cell destruction in pancreata from diabetes-prone NOD and non-diabetes-prone congenic NOD.H-2b mice between 3 and 16 weeks of age. RESULTS: Together with results showing the spatial dynamics of the insulitis process, we provide data of beta-cell volume distributions down to the level of the individual islets and throughout the pancreas during the development and progression of type 1 diabetes. Our data provide evidence for a compensatory growth potential of the larger insulin(+) islets during the later stages of the disease around the time point for development of clinical diabetes. This is in contrast to smaller islets, which appear less resistant to the autoimmune attack. We also provide new information on the spatial dynamics of the insulitis process itself, including its apparently random distribution at onset, the local variations during its further development, and the formation of structures resembling tertiary lymphoid organs at later phases of insulitis progression. CONCLUSIONS: Our data provide a powerful tool for phenotypic analysis of genetic and environmental effects on type 1 diabetes etiology as well as for evaluating the potential effect of therapeutic regimes.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Pancreas/pathology , Animals , Cell Size , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Disease Progression , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Insulin-Secreting Cells/immunology , Mice , Mice, Inbred NOD , Organ Size/immunology , Pancreas/immunology , Statistics, Nonparametric
14.
Islets ; 1(2): 163-4, 2009.
Article in English | MEDLINE | ID: mdl-21099267

ABSTRACT

The capacity to record the spatial and quantitative distribution of cellular subtypes involved in diabetogenic processes is a key element in experimental diabetes research. A non-invasive technique to accurately monitor parameters such as pancreatic ß-cell mass (BCM) and its distribution would provide a stepping stone in understanding different aspects of diabetes pathogenesis. It would also assist in the development of therapeutic regimes by providing a tool for the evaluation of anti-diabetic drugs or other curative or diagnostic measures. At present, a range of imaging modalities are being explored for this purpose. Whereas nuclear imaging techniques, characterised by their high tissue penetration depth but relatively low spatial resolution, appear most promising for the study of humans and large animals, optical imaging enables a route to cost-effective, high sensitivity, high resolution imaging in rodent models for disease. In this commentary, the potential impact of infrared fluorescent proteins (IFPs), as recently reported by Shu et al in Science, for imaging of the pancreas in small animals will be discussed.


Subject(s)
Diagnostic Imaging/trends , Fluorescent Dyes , Infrared Rays , Insulin-Secreting Cells/cytology , Luminescent Proteins , Animals , Diagnostic Imaging/methods , Humans , Insulin-Secreting Cells/metabolism , Mice , Optics and Photonics/methods , Pancreas/metabolism , Spectrophotometry, Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...