Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Diabetes Obes Metab ; 26(5): 1731-1745, 2024 May.
Article in English | MEDLINE | ID: mdl-38351663

ABSTRACT

AIM: Acyl-coenzyme A dehydrogenase family member 10 (ACAD10) is a mitochondrial protein purported to be involved in the fatty acid oxidation pathway. Metformin is the most prescribed therapy for type 2 diabetes; however, its precise mechanisms of action(s) are still being uncovered. Upregulation of ACAD10 is a requirement for metformin's ability to inhibit growth in cancer cells and extend lifespan in Caenorhabditis elegans. However, it is unknown whether ACAD10 plays a role in metformin's metabolic actions. MATERIALS AND METHODS: We assessed the role for ACAD10 on whole-body metabolism and metformin action by generating ACAD10KO mice on a C57BL/6J background via CRISPR-Cas9 technology. In-depth metabolic phenotyping was conducted in both sexes on a normal chow and high fat-high sucrose diet. RESULTS: Compared with wildtype mice, we detected no difference in body composition, energy expenditure or glucose tolerance in male or female ACAD10KO mice, on a chow diet or high-fat, high-sucrose diet (p ≥ .05). Hepatic mitochondrial function and insulin signalling was not different between genotypes under basal or insulin-stimulated conditions (p ≥ .05). Glucose excursions following acute administration of metformin before a glucose tolerance test were not different between genotypes nor was body composition or energy expenditure altered after 4 weeks of daily metformin treatment (p ≥ .05). Despite the lack of a metabolic phenotype, liver lipidomic analysis suggests ACAD10 depletion influences the abundance of specific ceramide species containing very long chain fatty acids, while metformin treatment altered clusters of cholesterol ester, plasmalogen, phosphatidylcholine and ceramide species. CONCLUSIONS: Loss of ACAD10 does not alter whole-body metabolism or impact the acute or chronic metabolic actions of metformin in this model.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Male , Female , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Mice, Inbred C57BL , Metformin/pharmacology , Glucose/metabolism , Insulin , Ceramides , Sucrose , Diet, High-Fat/adverse effects
2.
Cell Death Differ ; 30(6): 1447-1456, 2023 06.
Article in English | MEDLINE | ID: mdl-36894688

ABSTRACT

Many lymphoid malignancies arise from deregulated c-MYC expression in cooperation with additional genetic lesions. While many of these cooperative genetic lesions have been discovered and their functions characterised, DNA sequence data of primary patient samples suggest that many more do exist. However, the nature of their contributions to c-MYC driven lymphomagenesis have not yet been investigated. We identified TFAP4 as a potent suppressor of c-MYC driven lymphoma development in a previous genome-wide CRISPR knockout screen in primary cells in vivo [1]. CRISPR deletion of TFAP4 in Eµ-MYC transgenic haematopoietic stem and progenitor cells (HSPCs) and transplantation of these manipulated HSPCs into lethally irradiated animals significantly accelerated c-MYC-driven lymphoma development. Interestingly, TFAP4 deficient Eµ-MYC lymphomas all arose at the pre-B cell stage of B cell development. This observation prompted us to characterise the transcriptional profile of pre-B cells from pre-leukaemic mice transplanted with Eµ-MYC/Cas9 HSPCs that had been transduced with sgRNAs targeting TFAP4. This analysis revealed that TFAP4 deletion reduced expression of several master regulators of B cell differentiation, such as Spi1, SpiB and Pax5, which are direct target genes of both TFAP4 and MYC. We therefore conclude that loss of TFAP4 leads to a block in differentiation during early B cell development, thereby accelerating c-MYC-driven lymphoma development.


Subject(s)
Lymphoma , Proto-Oncogene Proteins c-myc , Mice , Animals , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Genes, myc , Lymphoma/pathology , Precursor Cells, B-Lymphoid/metabolism , Mice, Transgenic
4.
Nat Commun ; 13(1): 4739, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35961968

ABSTRACT

CRISPR technologies have advanced cancer modelling in mice, but CRISPR activation (CRISPRa) methods have not been exploited in this context. We establish a CRISPRa mouse (dCas9a-SAMKI) for inducing gene expression in vivo and in vitro. Using dCas9a-SAMKI primary lymphocytes, we induce B cell restricted genes in T cells and vice versa, demonstrating the power of this system. There are limited models of aggressive double hit lymphoma. Therefore, we transactivate pro-survival BCL-2 in Eµ-MycT/+;dCas9a-SAMKI/+ haematopoietic stem and progenitor cells. Mice transplanted with these cells rapidly develop lymphomas expressing high BCL-2 and MYC. Unlike standard Eµ-Myc lymphomas, BCL-2 expressing lymphomas are highly sensitive to the BCL-2 inhibitor venetoclax. We perform genome-wide activation screens in these lymphoma cells and find a dominant role for the BCL-2 protein A1 in venetoclax resistance. Here we show the potential of our CRISPRa model for mimicking disease and providing insights into resistance mechanisms towards targeted therapies.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Lymphoma , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sulfonamides
5.
Essays Biochem ; 66(1): 11-18, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35416243

ABSTRACT

Complex biomolecular technologies revolutionise scientific research. Fully embedding scientific advances in the community requires innovative ways to educate learners on the molecular foundations upon which these technologies are based. In this case study, we present the conception and design of Walter and Eliza Hall Institute of Medical Research (WEHI's) inaugural wholly online learning course focussed on explaining the revolutionary genome-editing technology, clustered regulatory interspaced palindromic repeats (CRISPR). Utilising WEHI's strength in bringing science educators and world-leading CRISPR scientists together, we designed a multimodal online resource that introduces learners, without an extensive background in either science or genome editing, to the fundamental concepts of CRISPR technology. Using the online course creation tool, Articulate 360, we guided learners through three modules containing targeted lessons designed to focus on specific learning outcomes. Integrated videos, research articles, interviews, and other resources, allowed for self-paced learning that met various learning style needs. The extensive resources provided opportunities to delve deeper into the content for advanced learners. The effectiveness of the course, evaluated with survey responses collected upon completion of the course, highlighted the ease of use and functionality of the course, and an increased understanding of CRISPR technology after course completion. We anticipate future online learning course development to showcase complex molecular technology that will be valuable for tertiary education, as well as for those in the wider community interested in understanding important advances in biomedicine.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Education, Distance , Gene Editing , Learning , Technology
6.
Cell Rep ; 34(11): 108851, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730574

ABSTRACT

Devil facial tumor disease (DFTD) and its lack of available therapies are propelling the Tasmanian devil population toward extinction. This study demonstrates that cholesterol homeostasis and carbohydrate energy metabolism sustain the proliferation of DFTD cells in a cell-type-dependent manner. In addition, we show that the liver-X nuclear receptor-ß (LXRß), a major cholesterol cellular sensor, and its natural ligand 24S-hydroxycholesterol promote the proliferation of DFTD cells via a metabolic switch toward aerobic glycolysis. As a proof of concept of the role of cholesterol homeostasis on DFTD proliferation, we show that atorvastatin, an FDA-approved statin-drug subtype used against human cardiovascular diseases that inhibits cholesterol synthesis, shuts down DFTD energy metabolism and prevents tumor growth in an in vivo DFTD-xenograft model. In conclusion, we show that intervention against cholesterol homeostasis and carbohydrate-dependent energy metabolism by atorvastatin constitutes a feasible biochemical treatment against DFTD, which may assist in the conservation of the Tasmanian devil.


Subject(s)
Cholesterol/metabolism , Facial Neoplasms/metabolism , Facial Neoplasms/veterinary , Homeostasis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver X Receptors/metabolism , Marsupialia/metabolism , Aerobiosis/drug effects , Animals , Atorvastatin/pharmacology , Cell Proliferation/drug effects , Facial Neoplasms/pathology , Female , Glycolysis/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Oxysterols/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Blood ; 137(5): 646-660, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33538798

ABSTRACT

Richter's transformation (RT) is an aggressive lymphoma that occurs upon progression from chronic lymphocytic leukemia (CLL). Transformation has been associated with genetic aberrations in the CLL phase involving TP53, CDKN2A, MYC, and NOTCH1; however, a significant proportion of RT cases lack CLL phase-associated events. Here, we report that high levels of AKT phosphorylation occur both in high-risk CLL patients harboring TP53 and NOTCH1 mutations as well as in patients with RT. Genetic overactivation of Akt in the murine Eµ-TCL1 CLL mouse model resulted in CLL transformation to RT with significantly reduced survival and an aggressive lymphoma phenotype. In the absence of recurrent mutations, we identified a profile of genomic aberrations intermediate between CLL and diffuse large B-cell lymphoma. Multiomics assessment by phosphoproteomic/proteomic and single-cell transcriptomic profiles of this Akt-induced murine RT revealed an S100 protein-defined subcluster of highly aggressive lymphoma cells that developed from CLL cells, through activation of Notch via Notch ligand expressed by T cells. Constitutively active Notch1 similarly induced RT of murine CLL. We identify Akt activation as an initiator of CLL transformation toward aggressive lymphoma by inducing Notch signaling between RT cells and microenvironmental T cells.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptor, Notch1/physiology , Animals , Clonal Evolution , Disease Progression , Enzyme Activation , Gene Expression Regulation, Neoplastic , Genes, p53 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/physiopathology , Mice , Mice, Inbred C57BL , Phenotype , Phosphoproteins/physiology , Proto-Oncogene Proteins c-akt/genetics , Receptors, Antigen, B-Cell/immunology , Signal Transduction/physiology , Transcriptome , Tumor Microenvironment , Tumor Suppressor Protein p53/physiology , Up-Regulation
8.
Biochem Soc Trans ; 49(1): 269-280, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33449100

ABSTRACT

CRISPR base editing technology is a promising genome editing tool as (i) it does not require a DNA template to introduce mutations and (ii) it avoids creating DNA double-strand breaks, which can lead to unintended chromosomal alterations or elicit an unwanted DNA damage response. Given many cancers originate from point mutations in cancer-driving genes, the application of base editing for either modelling tumour development, therapeutic editing, or functional screening is of great promise. In this review, we summarise current DNA base editing technologies and will discuss recent advancements and existing hurdles for its usage in cancer research.


Subject(s)
Cell Transformation, Neoplastic/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , DNA Mutational Analysis/methods , Genes, Neoplasm , Humans , Mutation , Neoplasms/genetics , Oncogenes/genetics
9.
Nature ; 574(7776): 63-68, 2019 10.
Article in English | MEDLINE | ID: mdl-31554967

ABSTRACT

The gp130 receptor cytokines IL-6 and CNTF improve metabolic homeostasis but have limited therapeutic use for the treatment of type 2 diabetes. Accordingly, we engineered the gp130 ligand IC7Fc, in which one gp130-binding site is removed from IL-6 and replaced with the LIF-receptor-binding site from CNTF, fused with the Fc domain of immunoglobulin G, creating a cytokine with CNTF-like, but IL-6-receptor-dependent, signalling. Here we show that IC7Fc improves glucose tolerance and hyperglycaemia and prevents weight gain and liver steatosis in mice. In addition, IC7Fc either increases, or prevents the loss of, skeletal muscle mass by activation of the transcriptional regulator YAP1. In human-cell-based assays, and in non-human primates, IC7Fc treatment results in no signs of inflammation or immunogenicity. Thus, IC7Fc is a realistic next-generation biological agent for the treatment of type 2 diabetes and muscle atrophy, disorders that are currently pandemic.


Subject(s)
Cytokine Receptor gp130/metabolism , Cytokines/chemical synthesis , Cytokines/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding, Competitive , Cytokines/chemistry , Diabetes Mellitus, Type 2/metabolism , Drug Design , Fatty Liver/prevention & control , Glucose Tolerance Test , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Incretins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Male , Mice , Muscle, Skeletal/drug effects , Obesity/metabolism , Pancreas/metabolism , Phosphoproteins/metabolism , Protein Engineering , Receptors, Interleukin-6/metabolism , Signal Transduction , Transcription Factors , Weight Gain/drug effects , YAP-Signaling Proteins
10.
Am J Physiol Endocrinol Metab ; 317(4): E597-E604, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31386565

ABSTRACT

It has been suggested that interleukin-6 (IL-6) produced by adipocytes in obesity leads to liver insulin resistance, although this hypothesis has never been definitively tested. Accordingly, we did so by generating adipocyte-specific IL-6-deficient (AdipoIL-6-/-) mice and studying them in the context of diet-induced and genetic obesity. Mice carrying two floxed alleles of IL-6 (C57Bl/6J) were crossed with Cre recombinase-overexpressing mice driven by the adiponectin promoter to generate AdipoIL-6-/- mice. AdipoIL-6-/- and floxed littermate controls were fed a standard chow or high-fat diet (HFD) for 16 wk and comprehensively metabolically phenotyped. In addition to a diet-induced obesity model, we also examined the role of adipocyte-derived IL-6 in a genetic model of obesity and insulin resistance by crossing the AdipoIL-6-/- mice with leptin-deficient (ob/ob) mice. As expected, mice on HFD and ob/ob mice displayed marked weight gain and increased fat mass compared with chow-fed and ob/+ (littermate control) animals, respectively. However, deletion of IL-6 from adipocytes in either model had no effect on glucose tolerance or fasting hyperinsulinemia. We concluded that adipocyte-specific IL-6 does not contribute to whole body glucose intolerance in obese mice.


Subject(s)
Adipocytes/metabolism , Glucose Intolerance/genetics , Interleukin-6/genetics , Obesity/genetics , Weight Gain/genetics , Adiponectin/biosynthesis , Adiponectin/genetics , Adiposity/genetics , Animals , Body Composition/genetics , Diet, High-Fat , Glucose Intolerance/etiology , Insulin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Obesity/metabolism
11.
Cell Death Differ ; 26(5): 902-917, 2019 05.
Article in English | MEDLINE | ID: mdl-30042493

ABSTRACT

BCL2A1 is an anti-apoptotic member of the BCL-2 family that contributes to chemoresistance in a subset of tumors. BCL2A1 has a short half-life due to its constitutive processing by the ubiquitin-proteasome system. This constitutes a major tumor-suppressor mechanism regulating BCL2A1 function. However, the enzymes involved in the regulation of BCL2A1 protein stability are currently unknown. Here, we provide the first insight into the regulation of BCL2A1 ubiquitination. We present evidence that TRIM28 is an E3 ubiquitin-ligase for BCL2A1. Indeed, endogenous TRIM28 and BCL2A1 bind to each other at the mitochondria and TRIM28 knock-down decreases BCL2A1 ubiquitination. We also show that TRIM17 stabilizes BCL2A1 by blocking TRIM28 from binding and ubiquitinating BCL2A1, and that GSK3 is involved in the phosphorylation-mediated inhibition of BCL2A1 degradation. BCL2A1 and its close relative MCL1 are thus regulated by common factors but with opposite outcome. Finally, overexpression of TRIM28 or knock-out of TRIM17 reduced BCLA1 protein levels and restored sensitivity of melanoma cells to BRAF-targeted therapy. Therefore, our data describe a molecular rheostat in which two proteins of the TRIM family antagonistically regulate BCL2A1 stability and modulate cell death.


Subject(s)
Apoptosis/genetics , Minor Histocompatibility Antigens/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif-Containing Protein 28/genetics , Ubiquitin-Protein Ligases/genetics , Cell Death/genetics , Cell Line, Tumor , Doxycycline/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3/genetics , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation/genetics , Proteasome Endopeptidase Complex/genetics , Protein Binding/genetics , Protein Stability , Proteolysis/drug effects , Ubiquitination/genetics
13.
J Physiol ; 594(2): 267-79, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26608096

ABSTRACT

Obesity is currently at epidemic levels worldwide and is associated with a wide range of diseases such as type 2 diabetes, cardiovascular disease, fatty liver disease and certain forms of cancer. Obesity-induced chronic inflammation is central to the disrupted metabolic homeostasis which underlies many of these conditions. While research over the past decade has identified many of the cells and signalling molecules that contribute to obesity-induced inflammation, perhaps the best characterised are the stress-activated c-Jun NH2 -terminal kinases (JNKs). JNKs are activated in obesity in numerous metabolically important cells and tissues such as adipose tissue, macrophages, liver, skeletal muscle and regions of the brain and pituitary. Elegant in vivo mouse studies using Cre-LoxP-mediated recombination of the JNK1 and JNK2 genes have revealed the remarkably diverse roles that JNKs play in the development of obesity-induced inflammation, impaired glucose homeostasis and hepatic steatosis. While JNK activation in classical metabolically active tissues such as skeletal muscle and adipose tissue only appears to play a minor role on the induction of the above-mentioned pathologies, recent studies have clearly established the important roles JNK signalling fulfils in macrophages, the liver and cells of the anterior pituitary. Collectively, these studies place JNKs as important mediators of obesity and obesity-associated disruptions to metabolic homeostasis.


Subject(s)
Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Obesity/metabolism , Animals , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Second Messenger Systems
14.
Cell Metab ; 21(5): 667-77, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25955204

ABSTRACT

Aging is a progressive decline of body function, during which many tissues accumulate few cells with high levels of deleted mitochondrial DNA (mtDNA), leading to a defect of mitochondrial functions. Whether this mosaic mitochondrial deficiency contributes to organ dysfunction is unknown. To investigate this, we generated mice with an accelerated accumulation of mtDNA deletions in the myocardium, by expressing a dominant-negative mutant mitochondrial helicase. These animals accumulated few randomly distributed cardiomyocytes with compromised mitochondrial function, which led to spontaneous ventricular premature contractions and AV blocks at 18 months. These symptoms were not caused by a general mitochondrial dysfunction in the entire myocardium, and were not observed in mice at 12 months with significantly lower numbers of dysfunctional cells. Therefore, our results suggest that the disposition to arrhythmia typically found in the aged human heart might be due to the random accumulation of mtDNA deletions and the subsequent mosaic respiratory chain deficiency.


Subject(s)
Aging , Arrhythmias, Cardiac/etiology , DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Oxygen/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Cell Respiration , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Deletion , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
15.
Cell Rep ; 9(4): 1495-506, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456138

ABSTRACT

Activation of c-Jun N-terminal kinase 1 (JNK1)- and inhibitor of nuclear factor kappa-B kinase 2 (IKK2)-dependent signaling plays a crucial role in the development of obesity-associated insulin and leptin resistance not only in peripheral tissues but also in the CNS. Here, we demonstrate that constitutive JNK activation in agouti-related peptide (AgRP)-expressing neurons of the hypothalamus is sufficient to induce weight gain and adiposity in mice as a consequence of hyperphagia. JNK activation increases spontaneous action potential firing of AgRP cells and causes both neuronal and systemic leptin resistance. Similarly, activation of IKK2 signaling in AgRP neurons also increases firing of these cells but fails to cause obesity and leptin resistance. In contrast to JNK activation, IKK2 activation blunts insulin signaling in AgRP neurons and impairs systemic glucose homeostasis. Collectively, these experiments reveal both overlapping and nonredundant effects of JNK- and IKK-dependent signaling in AgRP neurons, which cooperate in the manifestation of the metabolic syndrome.


Subject(s)
Agouti-Related Protein/metabolism , I-kappa B Kinase/metabolism , Insulin Resistance , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/enzymology , Obesity/enzymology , Action Potentials/drug effects , Adiposity/drug effects , Animals , Body Weight/drug effects , Enzyme Activation/drug effects , Glucose/metabolism , Homeostasis/drug effects , Insulin/metabolism , Leptin/pharmacology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Mutant Proteins/metabolism , Neurons/drug effects
16.
Immunol Cell Biol ; 92(4): 331-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24751614

ABSTRACT

The lack of physical activity and overnutrition in our modern lifestyle culminates in what we now experience as the current obesity and diabetes pandemic. Medical research performed over the past 20 years identified chronic low-grade inflammation as a mediator of these metabolic disorders. Hence, finding therapeutic strategies against this underlying inflammation and identifying molecules implicated in this process is of significant importance. Following the observation of an increased plasma concentration of interleukin-6 (IL-6) in obese patients, this protein, known predominantly as a pro-inflammatory cytokine, came into focus. In an attempt to clarify its importance, several studies implicated IL-6 as a co-inducer of the development of obesity-associated insulin resistance, which precedes the development of type 2 diabetes. However, the identification of IL-6 as a myokine, a protein produced and secreted by skeletal muscle to fulfil paracrine or endocrine roles in the insulin-sensitizing effects following exercise, provides a contrasting and hence paradoxical identity of this protein in the context of metabolism. We review here the literature considering the complex, pleiotropic role of IL-6 in the context of metabolism in health and disease.


Subject(s)
Interleukin-6/metabolism , Metabolism , Animals , Exercise , Humans , Immunity , Insulin Resistance , Interleukin-6/immunology , Metabolism/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/immunology , Obesity/metabolism , Obesity/pathology
17.
PLoS One ; 8(1): e54247, 2013.
Article in English | MEDLINE | ID: mdl-23349837

ABSTRACT

Obesity and associated metabolic disturbances, such as increased circulating fatty acids cause prolonged low grade activation of inflammatory signaling pathways in liver, skeletal muscle, adipose tissue and even in the CNS. Activation of inflammatory pathways in turn impairs insulin signaling, ultimately leading to obesity-associated type 2 diabetes mellitus. Conventional JNK-1 knock out mice are protected from high fat diet-induced insulin resistance, characterizing JNK-1-inhibition as a potential approach to improve glucose metabolism in obese patients. However, the cell type-specific role of elevated JNK-1 signaling as present during the course of obesity has not been fully elucidated yet. To investigate the functional contribution of altered JNK-1 activation in skeletal muscle, we have generated a ROSA26 insertion mouse strain allowing for Cre-activatable expression of a JNK-1 constitutive active construct (JNK(C)). To examine the consequence of skeletal muscle-restricted JNK-1 overactivation in the development of insulin resistance and glucose metabolism, JNK(C) mice were crossed to Mck-Cre mice yielding JNK(SM-C) mice. However, despite increased muscle-specific JNK activation, energy homeostasis and glucose metabolism in JNK(SM-C) mice remained largely unaltered compared to controls. In line with these findings, obese mice with skeletal muscle specific disruption of JNK-1, did not affect energy and glucose homeostasis. These experiments indicate that JNK-1 activation in skeletal muscle does not account for the major effects on diet-induced, JNK-1-mediated deterioration of insulin action and points towards a so far underappreciated role of JNK-1 in other tissues than skeletal muscle during the development of obesity-associated insulin resistance.


Subject(s)
Glucose/metabolism , Homeostasis , Insulin Resistance , Mitogen-Activated Protein Kinase 8/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Animals , Blotting, Western , Body Composition , Diet, High-Fat/adverse effects , Energy Metabolism , Female , Gene Expression , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/genetics , Obesity/etiology , Obesity/genetics , Phosphorylation , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Untranslated , Reverse Transcriptase Polymerase Chain Reaction
18.
J Biol Chem ; 287(14): 10771-9, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22351769

ABSTRACT

Exercise increases the expression of the prototypical myokine IL-6, but the precise mechanism by which this occurs has yet to be identified. To mimic exercise conditions, C2C12 myotubes were mechanically stimulated via electrical pulse stimulation (EPS). We compared the responses of EPS with the pharmacological Ca(2+) carrier calcimycin (A23187) because contraction induces marked increases in cytosolic Ca(2+) levels or the classical IκB kinase/NFκB inflammatory response elicited by H(2)O(2). We demonstrate that, unlike H(2)O(2)-stimulated increases in IL-6 mRNA, neither calcimycin- nor EPS-induced IL-6 mRNA expression is under the transcriptional control of NFκB. Rather, we show that EPS increased the phosphorylation of JNK and the reporter activity of the downstream transcription factor AP-1. Furthermore, JNK inhibition abolished the EPS-induced increase in IL-6 mRNA and protein expression. Finally, we observed an exercise-induced increase in both JNK phosphorylation and IL-6 mRNA expression in the skeletal muscles of mice after 30 min of treadmill running. Importantly, exercise did not increase IL-6 mRNA expression in skeletal muscle-specific JNK-deficient mice. These data identify a novel contraction-mediated transcriptional regulatory pathway for IL-6 in skeletal muscle.


Subject(s)
Interleukin-6/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , Animals , Calcimycin/pharmacology , Cell Line , Electric Stimulation , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects
19.
J Exp Med ; 208(9): 1749-56, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21844204

ABSTRACT

The substitution of one amino acid in the Roquin protein by the sanroque mutation induces a dramatic autoimmune syndrome in mice. This is believed to occur through ectopic expression of inducible T cell co-stimulator (ICOS) and unrestrained differentiation of follicular T helper cells, which induce spontaneous germinal center reactions to self-antigens. In this study, we demonstrate that tissue-specific ablation of Roquin in T or B cells, in the entire hematopoietic system, or in epithelial cells of transplanted thymi did not cause autoimmunity. Loss of Roquin induced elevated expression of ICOS through T cell-intrinsic and -extrinsic mechanisms, which itself was not sufficient to break self-tolerance. Instead, ablation of Roquin in the hematopoietic system caused defined changes in immune homeostasis, including the expansion of macrophages, eosinophils, and T cell subsets, most dramatically CD8 effector-like T cells, through cell-autonomous and nonautonomous mechanisms. Germline Roquin deficiency led to perinatal lethality, which was partially rescued on the genetic background of an outbred strain. However, not even complete absence of Roquin resulted in overt self-reactivity, suggesting that the sanroque mutation induces autoimmunity through an as yet unknown mechanism.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity , Gene Expression Regulation/immunology , Immunity, Cellular , Ubiquitin-Protein Ligases/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Gene Expression Regulation/genetics , Germinal Center/immunology , Germinal Center/metabolism , Inducible T-Cell Co-Stimulator Protein , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Proc Natl Acad Sci U S A ; 107(13): 6028-33, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231445

ABSTRACT

c-Jun N-terminal kinase (JNK) 1-dependent signaling plays a crucial role in the development of obesity-associated insulin resistance. Here we demonstrate that JNK activation not only occurs in peripheral tissues, but also in the hypothalamus and pituitary of obese mice. To resolve the importance of JNK1 signaling in the hypothalamic/pituitary circuitry, we have generated mice with a conditional inactivation of JNK1 in nestin-expressing cells (JNK1(DeltaNES) mice). JNK1(DeltaNES) mice exhibit improved insulin sensitivity both in the CNS and in peripheral tissues, improved glucose metabolism, as well as protection from hepatic steatosis and adipose tissue dysfunction upon high-fat feeding. Moreover, JNK1(DeltaNES) mice also show reduced somatic growth in the presence of reduced circulating growth hormone (GH) and insulin-like growth factor 1 (IGF1) concentrations, as well as increased thyroid axis activity. Collectively, these experiments reveal an unexpected, critical role for hypothalamic/pituitary JNK1 signaling in the coordination of metabolic/endocrine homeostasis.


Subject(s)
Glucose/metabolism , Hypothalamus/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Pituitary Gland/metabolism , Adiposity/physiology , Animals , Body Weight/physiology , Dietary Fats/administration & dosage , Growth Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Intermediate Filament Proteins/metabolism , Mice , Mice, Obese , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Nerve Tissue Proteins/metabolism , Nestin , Signal Transduction , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL