Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 222(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-36880935

ABSTRACT

Talin-1 is the core mechanosensitive adapter protein linking integrins to the cytoskeleton. The TLN1 gene is comprised of 57 exons that encode the 2,541 amino acid TLN1 protein. TLN1 was previously considered to be expressed as a single isoform. However, through differential pre-mRNA splicing analysis, we discovered a cancer-enriched, non-annotated 51-nucleotide exon in TLN1 between exons 17 and 18, which we refer to as exon 17b. TLN1 is comprised of an N-terminal FERM domain, linked to 13 force-dependent switch domains, R1-R13. Inclusion of exon 17b introduces an in-frame insertion of 17 amino acids immediately after Gln665 in the region between R1 and R2 which lowers the force required to open the R1-R2 switches potentially altering downstream mechanotransduction. Biochemical analysis of this isoform revealed enhanced vinculin binding, and cells expressing this variant show altered adhesion dynamics and motility. Finally, we showed that the TGF-ß/SMAD3 signaling pathway regulates this isoform switch. Future studies will need to consider the balance of these two TLN1 isoforms.


Subject(s)
Neoplasms , Talin , Humans , Talin/genetics , Mechanotransduction, Cellular , Exons/genetics , Adaptor Proteins, Signal Transducing
2.
Front Bioinform ; 2: 786898, 2022.
Article in English | MEDLINE | ID: mdl-36304260

ABSTRACT

RNA-seq analysis of alternative pre-mRNA splicing has facilitated an unprecedented understanding of transcriptome complexity in health and disease. However, despite the availability of countless bioinformatic pipelines for transcriptome-wide splicing analysis, the use of these tools is often limited to expert bioinformaticians. The need for high computational power, combined with computational outputs that are complicated to visualize and interpret present obstacles to the broader research community. Here we introduce DJExpress, an R package for differential expression analysis of transcriptomic features and expression-trait associations. To determine gene-level differential junction usage as well as associations between junction expression and molecular/clinical features, DJExpress uses raw splice junction counts as input data. Importantly, DJExpress runs on an average laptop computer and provides a set of interactive and intuitive visualization formats. In contrast to most existing pipelines, DJExpress can handle both annotated and de novo identified splice junctions, thereby allowing the quantification of novel splice events. Moreover, DJExpress offers a web-compatible graphical interface allowing the analysis of user-provided data as well as the visualization of splice events within our custom database of differential junction expression in cancer (DJEC DB). DJEC DB includes not only healthy and tumor tissue junction expression data from TCGA and GTEx repositories but also cancer cell line data from the DepMap project. The integration of DepMap functional genomics data sets allows association of junction expression with molecular features such as gene dependencies and drug response profiles. This facilitates identification of cancer cell models for specific splicing alterations that can then be used for functional characterization in the lab. Thus, DJExpress represents a powerful and user-friendly tool for exploration of alternative splicing alterations in RNA-seq data, including multi-level data integration of alternative splicing signatures in healthy tissue, tumors and cancer cell lines.

3.
Nat Commun ; 11(1): 4077, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796846

ABSTRACT

Double-strand breaks (DSBs) are the most toxic type of DNA lesions. Cells repair these lesions using either end protection- or end resection-coupled mechanisms. To study DSB repair choice, we present the Color Assay Tracing-Repair (CAT-R) to simultaneously quantify DSB repair via end protection and end resection pathways. CAT-R introduces DSBs using CRISPR/Cas9 in a tandem fluorescent reporter, whose repair distinguishes small insertions/deletions from large deletions. We demonstrate CAT-R applications in chemical and genetic screens. First, we evaluate 21 compounds currently in clinical trials which target the DNA damage response. Second, we examine how 417 factors involved in DNA damage response influence the choice between end protection and end resection. Finally, we show that impairing nucleotide excision repair favors error-free repair, providing an alternative way for improving CRISPR/Cas9-based knock-ins. CAT-R is a high-throughput, versatile assay to assess DSB repair choice, which facilitates comprehensive studies of DNA repair and drug efficiency testing.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Breaks, Double-Stranded , DNA Repair , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle , Cell Survival , DNA Damage , DNA End-Joining Repair , Drug Evaluation, Preclinical , Gene Knockout Techniques , HEK293 Cells , Humans , Poly (ADP-Ribose) Polymerase-1/genetics
4.
Nat Chem Biol ; 15(4): 340-347, 2019 04.
Article in English | MEDLINE | ID: mdl-30778204

ABSTRACT

Small nuclear RNAs (snRNAs) are core spliceosome components and mediate pre-mRNA splicing. Here we show that snRNAs contain a regulated and reversible nucleotide modification causing them to exist as two different methyl isoforms, m1 and m2, reflecting the methylation state of the adenosine adjacent to the snRNA cap. We find that snRNA biogenesis involves the formation of an initial m1 isoform with a single-methylated adenosine (2'-O-methyladenosine, Am), which is then converted to a dimethylated m2 isoform (N6,2'-O-dimethyladenosine, m6Am). The relative m1 and m2 isoform levels are determined by the RNA demethylase FTO, which selectively demethylates the m2 isoform. We show FTO is inhibited by the oncometabolite D-2-hydroxyglutarate, resulting in increased m2-snRNA levels. Furthermore, cells that exhibit high m2-snRNA levels show altered patterns of alternative splicing. Together, these data reveal that FTO controls a previously unknown central step of snRNA processing involving reversible methylation, and suggest that epitranscriptomic information in snRNA may influence mRNA splicing.


Subject(s)
Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/physiology , RNA, Small Nuclear/biosynthesis , Adenosine/biosynthesis , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alternative Splicing , Animals , HEK293 Cells , Humans , Male , Methylation , Mice , Mice, Knockout , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Nuclear/metabolism
5.
FEBS Lett ; 592(12): 2012-2022, 2018 06.
Article in English | MEDLINE | ID: mdl-29754392

ABSTRACT

The fate of mRNA is regulated by epitranscriptomic nucleotide modifications, the most abundant of which is N6 -methyladenosine (m6 A). Although the pattern and distribution of m6 A in mRNA is mediated by specific methyltransferases, a recent hypothesis is that specific demethylases or 'erasers' allow m6 A to be dynamically reversed by signaling pathways. In this Review, we discuss the data in support and against this model. New insights into the function of fat mass and obesity-associated protein (FTO), the original enzyme thought to be an m6 A eraser, reveal that its physiologic target is not m6 A, but instead is N6 ,2'-O-dimethyladenosine (m6 Am ). Another m6 A demethylase, ALKBH5, appears to have functions limited to sperm development in normal mice. Overall, the majority of the data suggest that m6 A is generally not reversible, although m6 A may be susceptible to demethylation in pathophysiological states such as cancer.


Subject(s)
Adenosine/analogs & derivatives , Epigenesis, Genetic , RNA, Messenger/chemistry , Adenosine/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Humans , RNA, Messenger/metabolism
6.
Sci Signal ; 11(527)2018 04 24.
Article in English | MEDLINE | ID: mdl-29692363

ABSTRACT

The transient activation of inflammatory networks is required for adipose tissue remodeling including the "browning" of white fat in response to stimuli such as ß3-adrenergic receptor activation. In this process, white adipose tissue acquires thermogenic characteristics through the recruitment of so-called beige adipocytes. We investigated the downstream signaling pathways impinging on adipocyte progenitors that promote de novo formation of adipocytes. We showed that the Jak family of kinases controlled TGFß signaling in the adipose tissue microenvironment through Stat3 and thereby adipogenic commitment, a function that was required for beige adipocyte differentiation of murine and human progenitors. Jak/Stat3 inhibited TGFß signaling to the transcription factors Srf and Smad3 by repressing local Tgfb3 and Tgfb1 expression before the core transcriptional adipogenic cascade was activated. This pathway cross-talk was triggered in stromal cells by ATGL-dependent adipocyte lipolysis and a transient wave of IL-6 family cytokines at the onset of adipose tissue remodeling induced by ß3-adrenergic receptor stimulation. Our results provide insight into the activation of adipocyte progenitors and are relevant for the therapeutic targeting of adipose tissue inflammatory pathways.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue/metabolism , Inflammation/genetics , Janus Kinases/genetics , Transforming Growth Factor beta/genetics , Adipocytes, Beige/pathology , Adipogenesis/genetics , Adipose Tissue/pathology , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Gene Expression Profiling , Humans , Inflammation/metabolism , Janus Kinases/metabolism , Lipase/genetics , Lipase/metabolism , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism
7.
Nat Commun ; 9(1): 1646, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695802

ABSTRACT

Colorectal cancer (CRC) is one of the most lethal cancers worldwide in which the vast majority of cases exhibit little genetic risk but are associated with a sedentary lifestyle and obesity. Although the mechanisms underlying CRC and colitis-associated colorectal cancer (CAC) remain unclear, we hypothesised that obesity-induced inflammation predisposes to CAC development. Here, we show that diet-induced obesity accelerates chemically-induced CAC in mice via increased inflammation and immune cell recruitment. Obesity-induced interleukin-6 (IL-6) shifts macrophage polarisation towards tumour-promoting macrophages that produce the chemokine CC-chemokine-ligand-20 (CCL-20) in the CAC microenvironment. CCL-20 promotes CAC progression by recruiting CC-chemokine-receptor-6 (CCR-6)-expressing B cells and γδ T cells via chemotaxis. Compromised cell recruitment as well as inhibition of B and γδ T cells protects against CAC progression. Collectively, our data reveal a function for IL-6 in the CAC microenvironment via lymphocyte recruitment through the CCL-20/CCR-6 axis, thereby implicating a potential therapeutic intervention for human patients.


Subject(s)
Chemokine CCL20/metabolism , Colitis, Ulcerative/pathology , Colorectal Neoplasms/immunology , Interleukin-6/metabolism , Obesity/immunology , Receptors, CCR6/metabolism , Animals , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Chemokine CCL20/immunology , Chemotaxis/immunology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Colon/drug effects , Colon/immunology , Colon/pathology , Colorectal Neoplasms/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Progression , Female , Humans , Interleukin-6 Receptor alpha Subunit/genetics , Interleukin-6 Receptor alpha Subunit/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Obesity/etiology , Receptors, CCR6/genetics , Signal Transduction/immunology , Tumor Microenvironment/immunology
8.
Cell Metab ; 26(1): 171-184.e6, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683285

ABSTRACT

Natural killer (NK) cells contribute to the development of obesity-associated insulin resistance. We demonstrate that in mice obesity promotes expansion of a distinct, interleukin-6 receptor (IL6R)a-expressing NK subpopulation, which also expresses a number of other myeloid lineage genes such as the colony-stimulating factor 1 receptor (Csf1r). Selective ablation of this Csf1r-expressing NK cell population prevents obesity and insulin resistance. Moreover, conditional inactivation of IL6Ra or Stat3 in NK cells limits obesity-associated formation of these myeloid signature NK cells, protecting from obesity, insulin resistance, and obesity-associated inflammation. Also in humans IL6Ra+ NK cells increase in obesity and correlate with markers of systemic low-grade inflammation, and their gene expression profile overlaps with characteristic gene sets of NK cells in obese mice. Collectively, we demonstrate that obesity-associated inflammation and metabolic disturbances depend on interleukin-6/Stat3-dependent formation of a distinct NK population, which may provide a target for the treatment of obesity, metaflammation-associated pathologies, and diabetes.


Subject(s)
Energy Metabolism , Glucose/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Killer Cells, Natural/pathology , Obesity/metabolism , STAT3 Transcription Factor/metabolism , Adult , Animals , Homeostasis , Humans , Inflammation/complications , Inflammation/pathology , Insulin Resistance , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , Young Adult
9.
J Immunol ; 198(7): 2927-2934, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28193830

ABSTRACT

Obesity is associated with chronic low-grade inflammation of adipose tissue (AT) and an increase of AT macrophages (ATMs) that is linked to the onset of type 2 diabetes. We have recently shown that focal sites of inflammation around dying adipocytes, so-called crown-like structures, exhibit a unique microenvironment for macrophage proliferation. Interestingly, locally proliferating macrophages were not classically activated (M1), but they exhibited a rather alternatively activated (M2) immune phenotype. In this study, we established organotypic cell cultures of AT explants to study the impact of cytokine treatment on local ATM proliferation, without the bias of early monocyte recruitment. We show that exposure of AT to Th2 cytokines, such as IL-4, IL-13, and GM-CSF, stimulates ATM proliferation, whereas Th1 cytokines, such as TNF-α, inhibit local ATM proliferation. Furthermore, AT from obese mice exhibits an increased sensitivity to IL-4 stimulation, indicated by an increased phosphorylation of STAT6. In line with this, gene expression of the IL-4 receptor (Il4ra) and its ligand IL-13 are elevated in AT of obese C57BL/6 mice. Most importantly, Il4ra expression and susceptibility to IL-4 or IL-13 treatment depend on IL-6 signaling, which seems to be the underlying mechanism of local ATM proliferation in obesity. We conclude that IL-6 acts as a Th2 cytokine in obesity by stimulating M2 polarization and local ATM proliferation, presumably due to upregulation of the IL-4 receptor α.


Subject(s)
Adipose Tissue/immunology , Cell Proliferation , Interleukin-6/immunology , Macrophages/immunology , Obesity/immunology , Animals , Blotting, Western , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/immunology , Disease Models, Animal , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Transcriptome
10.
Soc Cogn Affect Neurosci ; 12(4): 618-634, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28119508

ABSTRACT

Social learning is fundamental to human interactions, yet its computational and physiological mechanisms are not well understood. One prominent open question concerns the role of neuromodulatory transmitters. We combined fMRI, computational modelling and genetics to address this question in two separate samples (N = 35, N = 47). Participants played a game requiring inference on an adviser's intentions whose motivation to help or mislead changed over time. Our analyses suggest that hierarchically structured belief updates about current advice validity and the adviser's trustworthiness, respectively, depend on different neuromodulatory systems. Low-level prediction errors (PEs) about advice accuracy not only activated regions known to support 'theory of mind', but also the dopaminergic midbrain. Furthermore, PE responses in ventral striatum were influenced by the Met/Val polymorphism of the Catechol-O-Methyltransferase (COMT) gene. By contrast, high-level PEs ('expected uncertainty') about the adviser's fidelity activated the cholinergic septum. These findings, replicated in both samples, have important implications: They suggest that social learning rests on hierarchically related PEs encoded by midbrain and septum activity, respectively, in the same manner as other forms of learning under volatility. Furthermore, these hierarchical PEs may be broadcast by dopaminergic and cholinergic projections to induce plasticity specifically in cortical areas known to represent beliefs about others.


Subject(s)
Dopamine/physiology , Mesencephalon/physiology , Septum Pellucidum/physiology , Social Learning/physiology , Adult , Brain Mapping , Catechol O-Methyltransferase/genetics , Culture , Dopamine/genetics , Humans , Intention , Magnetic Resonance Imaging , Male , Motivation/physiology , Polymorphism, Single Nucleotide/genetics , Tyrosine 3-Monooxygenase/genetics , Young Adult
11.
Nature ; 541(7637): 371-375, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28002401

ABSTRACT

Internal bases in mRNA can be subjected to modifications that influence the fate of mRNA in cells. One of the most prevalent modified bases is found at the 5' end of mRNA, at the first encoded nucleotide adjacent to the 7-methylguanosine cap. Here we show that this nucleotide, N6,2'-O-dimethyladenosine (m6Am), is a reversible modification that influences cellular mRNA fate. Using a transcriptome-wide map of m6Am we find that m6Am-initiated transcripts are markedly more stable than mRNAs that begin with other nucleotides. We show that the enhanced stability of m6Am-initiated transcripts is due to resistance to the mRNA-decapping enzyme DCP2. Moreover, we find that m6Am is selectively demethylated by fat mass and obesity-associated protein (FTO). FTO preferentially demethylates m6Am rather than N6-methyladenosine (m6A), and reduces the stability of m6Am mRNAs. Together, these findings show that the methylation status of m6Am in the 5' cap is a dynamic and reversible epitranscriptomic modification that determines mRNA stability.


Subject(s)
Adenosine/analogs & derivatives , RNA Caps/chemistry , RNA Caps/metabolism , RNA Stability , Adenosine/chemistry , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Endoribonucleases/metabolism , Epigenesis, Genetic , Guanosine/analogs & derivatives , Guanosine/metabolism , HEK293 Cells , Half-Life , Humans , Male , Methylation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Substrate Specificity , Transcription Initiation Site , Transcriptome
13.
Cell ; 165(4): 882-95, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133169

ABSTRACT

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Subject(s)
Brain/metabolism , Diet, High-Fat , Glucose/metabolism , Obesity/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/metabolism , Cognition , Endothelial Cells/metabolism , Fatty Acids/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Myeloid Cells/metabolism
14.
Cell ; 165(1): 125-138, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27015310

ABSTRACT

Activation of Agouti-related peptide (AgRP) neurons potently promotes feeding, and chronically altering their activity also affects peripheral glucose homeostasis. We demonstrate that acute activation of AgRP neurons causes insulin resistance through impairment of insulin-stimulated glucose uptake into brown adipose tissue (BAT). AgRP neuron activation acutely reprograms gene expression in BAT toward a myogenic signature, including increased expression of myostatin. Interference with myostatin activity improves insulin sensitivity that was impaired by AgRP neurons activation. Optogenetic circuitry mapping reveals that feeding and insulin sensitivity are controlled by both distinct and overlapping projections. Stimulation of AgRP → LHA projections impairs insulin sensitivity and promotes feeding while activation of AgRP → anterior bed nucleus of the stria terminalis (aBNST)vl projections, distinct from AgRP → aBNSTdm projections controlling feeding, mediate the effect of AgRP neuron activation on BAT-myostatin expression and insulin sensitivity. Collectively, our results suggest that AgRP neurons in mice induce not only eating, but also insulin resistance by stimulating expression of muscle-related genes in BAT, revealing a mechanism by which these neurons rapidly coordinate hunger states with glucose homeostasis.


Subject(s)
Adipose Tissue, Brown/metabolism , Appetite Regulation , Glucose/metabolism , Insulin Resistance , Neurons/metabolism , Agouti-Related Protein/metabolism , Animals , Feeding Behavior , Mice , Myostatin/genetics , Optogenetics , Transcriptome
15.
Nat Cell Biol ; 18(3): 328-36, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26900752

ABSTRACT

Activation of brown adipose tissue (BAT) controls energy homeostasis in rodents and humans and has emerged as an innovative strategy for the treatment of obesity and type 2 diabetes mellitus. Here we show that ageing- and obesity-associated dysfunction of brown fat coincides with global microRNA downregulation due to reduced expression of the microRNA-processing node Dicer1. Consequently, heterozygosity of Dicer1 in BAT aggravated diet-induced-obesity (DIO)-evoked deterioration of glucose metabolism. Analyses of differential microRNA expression during preadipocyte commitment and mouse models of progeria, longevity and DIO identified miR-328 as a regulator of BAT differentiation. Reducing miR-328 blocked preadipocyte commitment, whereas miR-328 overexpression instigated BAT differentiation and impaired muscle progenitor commitment-partly through silencing of the ß-secretase Bace1. Loss of Bace1 enhanced brown preadipocyte specification in vitro and was overexpressed in BAT of obese and progeroid mice. In vivo Bace1 inhibition delayed DIO-induced weight gain and improved glucose tolerance and insulin sensitivity. These experiments reveal Dicer1-miR-328-Bace1 signalling as a determinant of BAT function, and highlight the potential of Bace1 inhibition as a therapeutic approach to improve not only neurodegenerative diseases but also ageing- and obesity-associated impairments of BAT function.


Subject(s)
Adipose Tissue, Brown/metabolism , Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Cell Differentiation/physiology , DEAD-box RNA Helicases/genetics , MicroRNAs/genetics , Ribonuclease III/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , DEAD-box RNA Helicases/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Insulin Resistance/physiology , Mice, Inbred C57BL , MicroRNAs/metabolism , Obesity/genetics , Obesity/metabolism , Ribonuclease III/metabolism
16.
Neuroimage ; 128: 21-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26767945

ABSTRACT

Variations in the fat mass and obesity associated (FTO) gene are currently the strongest known genetic factor predisposing humans to non-monogenic obesity. Recent experiments have linked these variants to a broad spectrum of behavioural alterations, including food choice and substance abuse. Yet, the underlying neurobiological mechanisms by which these genetic variations influence body weight remain elusive. Here, we explore the brain structural substrate of the obesity-predisposing rs9939609 T/A variant of the FTO gene in non-obese subjects by means of multivariate classification and use fMRI to investigate genotype-specific differences in neural food-cue reactivity by analysing correlates of a visual food perception task. Our findings demonstrate that MRI-derived measures of morphology along middle and posterior fusiform gyrus (FFG) are highly predictive for FTO at-risk allele carriers, who also show enhanced neural responses elicited by food cues in the same posterior FFG area. In brief, these findings provide first-time evidence for FTO-specific differences in both brain structure and function already in non-obese individuals, thereby contributing to a mechanistic understanding of why FTO is a predisposing factor for obesity.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Obesity/genetics , Temporal Lobe/physiology , Visual Perception , Adult , Female , Food , Genetic Predisposition to Disease/genetics , Genotype , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Support Vector Machine
17.
J Neurosci ; 35(36): 12584-92, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26354923

ABSTRACT

Variations in the fat mass and obesity-associated (FTO) gene are linked to obesity. However, the underlying neurobiological mechanisms by which these genetic variants influence obesity, behavior, and brain are unknown. Given that Fto regulates D2/3R signaling in mice, we tested in humans whether variants in FTO would interact with a variant in the ANKK1 gene, which alters D2R signaling and is also associated with obesity. In a behavioral and fMRI study, we demonstrate that gene variants of FTO affect dopamine (D2)-dependent midbrain brain responses to reward learning and behavioral responses associated with learning from negative outcome in humans. Furthermore, dynamic causal modeling confirmed that FTO variants modulate the connectivity in a basic reward circuit of meso-striato-prefrontal regions, suggesting a mechanism by which genetic predisposition alters reward processing not only in obesity, but also in other disorders with altered D2R-dependent impulse control, such as addiction. Significance statement: Variations in the fat mass and obesity-associated (FTO) gene are associated with obesity. Here we demonstrate that variants of FTO affect dopamine-dependent midbrain brain responses and learning from negative outcomes in humans during a reward learning task. Furthermore, FTO variants modulate the connectivity in a basic reward circuit of meso-striato-prefrontal regions, suggesting a mechanism by which genetic vulnerability in reward processing can increase predisposition to obesity.


Subject(s)
Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Receptors, Dopamine D2/metabolism , Reward , Adult , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Connectome , Female , Humans , Male , Mesencephalon/metabolism , Mesencephalon/physiology
18.
Trends Immunol ; 36(2): 92-101, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25616716

ABSTRACT

Owing to its abundance in inflammatory settings, interleukin IL-6 is frequently viewed as a proinflammatory cytokine, with functions that parallel those of tumor necrosis factor (TNF) and IL-1ß in the context of inflammation. However, accumulating evidence points to a broader role for IL-6 in a variety of (patho)physiological conditions, including functions related to the resolution of inflammation. We review recent findings on the complex biological functions governed by IL-6 signaling, focusing on its role in inflammation-associated cancer and metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM). We propose that the anti-inflammatory functions of IL-6 may extend to multiple settings and cell types, and suggest that these dimensions should be incorporated in therapeutic approaches to these diseases. Finally, we outline important areas of inquiry towards understanding this pleiotropic cytokine.


Subject(s)
Interleukin-6/metabolism , Neoplasms/metabolism , Animals , Gene Expression Regulation , Humans , Liver/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Muscle, Skeletal/metabolism , Neoplasms/genetics , Neoplasms/immunology , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Signal Transduction
19.
Cell Metab ; 20(4): 678-86, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295788

ABSTRACT

Ceramides increase during obesity and promote insulin resistance. Ceramides vary in acyl-chain lengths from C14:0 to C30:0 and are synthesized by six ceramide synthase enzymes (CerS1-6). It remains unresolved whether obesity-associated alterations of specific CerSs and their defined acyl-chain length ceramides contribute to the manifestation of metabolic diseases. Here we reveal that CERS6 mRNA expression and C16:0 ceramides are elevated in adipose tissue of obese humans, and increased CERS6 expression correlates with insulin resistance. Conversely, CerS6-deficient (CerS6(Δ/Δ)) mice exhibit reduced C16:0 ceramides and are protected from high-fat-diet-induced obesity and glucose intolerance. CerS6 deletion increases energy expenditure and improves glucose tolerance, not only in CerS6(Δ/Δ) mice, but also in brown adipose tissue- (CerS6(ΔBAT)) and liver-specific (CerS6(ΔLIVER)) CerS6 knockout mice. CerS6 deficiency increases lipid utilization in BAT and liver. These experiments highlight CerS6 inhibition as a specific approach for the treatment of obesity and type 2 diabetes mellitus, circumventing the side effects of global ceramide synthesis inhibition.


Subject(s)
Ceramides/metabolism , Glucose Intolerance , Sphingosine N-Acyltransferase/metabolism , Adipose Tissue, Brown/metabolism , Animals , Body Mass Index , Diet, High-Fat , Female , Humans , Lipid Peroxidation , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/metabolism , Obesity/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Sphingosine N-Acyltransferase/deficiency , Sphingosine N-Acyltransferase/genetics , Weight Gain
20.
PLoS Genet ; 10(6): e1004385, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945157

ABSTRACT

Although mitochondrial dysfunction is often accompanied by excessive reactive oxygen species (ROS) production, we previously showed that an increase in random somatic mtDNA mutations does not result in increased oxidative stress. Normal levels of ROS and oxidative stress could also be a result of an active compensatory mechanism such as a mild increase in proton leak. Uncoupling protein 2 (UCP2) was proposed to play such a role in many physiological situations. However, we show that upregulation of UCP2 in mtDNA mutator mice is not associated with altered proton leak kinetics or ROS production, challenging the current view on the role of UCP2 in energy metabolism. Instead, our results argue that high UCP2 levels allow better utilization of fatty acid oxidation resulting in a beneficial effect on mitochondrial function in heart, postponing systemic lactic acidosis and resulting in longer lifespan in these mice. This study proposes a novel mechanism for an adaptive response to mitochondrial cardiomyopathy that links changes in metabolism to amelioration of respiratory chain deficiency and longer lifespan.


Subject(s)
Energy Metabolism/genetics , Fatty Acids/metabolism , Ion Channels/genetics , Mitochondria, Heart/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Acidosis, Lactic/metabolism , Animals , Cardiomyopathies/pathology , Eating/genetics , Life Expectancy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondrial Diseases/metabolism , Myocardium/metabolism , Oxidation-Reduction , Oxidative Stress , Proton Pumps/genetics , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL
...