Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(10): e23189, 2023 10.
Article in English | MEDLINE | ID: mdl-37713040

ABSTRACT

A protein altering variant in the gene encoding zinc finger homeobox-3 (ZFHX3) has recently been associated with lower BMI in a human genome-wide association study. We investigated metabolic parameters in mice harboring a missense mutation in Zfhx3 (Zfhx3Sci/+ ) and looked for altered in situ expression of transcripts that are associated with energy balance in the hypothalamus to understand how ZFHX3 may influence growth and metabolic effects. One-year-old male and female Zfhx3Sci/+ mice weighed less, had shorter body length, lower fat mass, smaller mesenteric fat depots, and lower circulating insulin, leptin, and insulin-like growth factor-1 (IGF1) concentrations than Zfhx3+/+ littermates. In a second cohort of 9-20-week-old males and females, Zfhx3Sci/+ mice ate less than wildtype controls, in proportion to body weight. In a third cohort of female-only Zfhx3Sci/+ and Zfhx3+/+ mice that underwent metabolic phenotyping from 6 to 14 weeks old, Zfhx3Sci/+ mice weighed less and had lower lean mass and energy expenditure, but fat mass did not differ. We detected increased expression of somatostatin and decreased expression of growth hormone-releasing hormone and growth hormone-receptor mRNAs in the arcuate nucleus (ARC). Similarly, ARC expression of orexigenic neuropeptide Y was decreased and ventricular ependymal expression of orphan G protein-coupled receptor Gpr50 was decreased. We demonstrate for the first time an energy balance effect of the Zfhx3Sci mutation, likely by altering expression of key ARC neuropeptides to alter growth, food intake, and energy expenditure.


Subject(s)
Genes, Homeobox , Homeodomain Proteins , Hypothalamus , Mutation, Missense , Animals , Female , Male , Mice , Gene Expression , Genome-Wide Association Study , Homeodomain Proteins/genetics , Hypothalamus/metabolism , Zinc Fingers
2.
ACS Med Chem Lett ; 11(12): 2421-2427, 2020 Dec 10.
Article in English | MEDLINE | ID: mdl-33335663

ABSTRACT

Utrophin modulation is a disease-modifying therapeutic strategy for Duchenne muscular dystrophy that would be applicable to all patient populations. To improve the suboptimal profile of ezutromid, the first-in-class clinical candidate, a second generation of utrophin modulators bearing a phosphinate ester moiety was developed. This modification significantly improved the physicochemical and ADME properties, but one of the main lead molecules was found to have dose-limiting hepatotoxicity. In this work we describe how less lipophilic analogues retained utrophin modulatory activity in a reporter gene assay, upregulated utrophin protein in dystrophic mouse muscle cells, but also had improved physicochemical and ADME properties. Notably, ClogP was found to directly correlate with pIC50 in HepG2 cells, hence leading to a potentially safer toxicological profiles in this series. Compound 21 showed a balanced profile (H2K EC50: 4.17 µM, solubility: 477 µM, mouse hepatocyte T 1/2 > 240 min) and increased utrophin protein 1.6-fold in a Western blot assay.

3.
Tetrahedron ; 76(2): 130819, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-32713969

ABSTRACT

Following on from ezutromid, the first-in-class benzoxazole utrophin modulator that progressed to Phase 2 clinical trials for the treatment of Duchenne muscular dystrophy, a new chemotype was designed to optimise its physicochemical and ADME profile. Herein we report the synthesis of SMT022357, a second generation utrophin modulator preclinical candidate, and an asymmetric synthesis of its constituent enantiomers. The pharmacological properties of both enantiomers were evaluated in vitro and in vivo. No significant difference in the activity or efficacy was observed between the two enantiomers; activity was found to be comparable to the racemic mixture.

4.
J Med Chem ; 63(14): 7880-7891, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32551645

ABSTRACT

Utrophin modulation is a promising therapeutic strategy for Duchenne muscular dystrophy (DMD), which should be applicable to all patient populations. Following on from ezutromid, the first-generation utrophin modulator, we describe the development of a second generation of utrophin modulators, based on the bioisosteric replacement of the sulfone group with a phosphinate ester and substitution of the metabolically labile naphthalene with a haloaryl substituent. The improved physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties, further reflected in the enhanced pharmacokinetic profile of the most advanced compounds, 30 and 27, led to significantly better in vivo exposure compared to ezutromid and alleviation of the dystrophic phenotype in mdx mice. While 30 was found to have dose-limiting hepatotoxicity, 27 and its enantiomers exhibited limited off-target effects, resulting in a safe profile and highlighting their potential utility as next-generation utrophin modulators suitable for progression toward a future DMD therapy.


Subject(s)
Benzoxazoles/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Utrophin/metabolism , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/pharmacokinetics , Benzoxazoles/toxicity , Escherichia coli/drug effects , Mice, Inbred mdx , Molecular Structure , Muscular Dystrophy, Duchenne/metabolism , Mutagenicity Tests , Rats , Salmonella typhimurium/drug effects , Stereoisomerism , Structure-Activity Relationship , Up-Regulation/drug effects
5.
Angew Chem Int Ed Engl ; 59(6): 2420-2428, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31755636

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.


Subject(s)
Benzoxazoles/chemistry , Naphthalenes/chemistry , Proteomics/methods , Receptors, Aryl Hydrocarbon/metabolism , Utrophin/metabolism , Animals , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Cycloaddition Reaction , Drug Design , Humans , Kinetics , Mice , Molecular Probes/chemistry , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/cytology , Myoblasts/metabolism , Naphthalenes/metabolism , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Protein Binding , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Up-Regulation/drug effects , Utrophin/agonists , Utrophin/genetics
6.
Hum Mol Genet ; 28(13): 2189-2200, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30990876

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal neuromuscular disorder caused by loss of dystrophin. Several therapeutic modalities are currently in clinical trials but none will achieve maximum functional rescue and full disease correction. Therefore, we explored the potential of combining the benefits of dystrophin with increases of utrophin, an autosomal paralogue of dystrophin. Utrophin and dystrophin can be co-expressed and co-localized at the same muscle membrane. Wild-type (wt) levels of dystrophin are not significantly affected by a moderate increase of utrophin whereas higher levels of utrophin reduce wt dystrophin, suggesting a finite number of actin binding sites at the sarcolemma. Thus, utrophin upregulation strategies may be applied to the more mildly affected Becker patients with lower dystrophin levels. Whereas increased dystrophin in wt animals does not offer functional improvement, overexpression of utrophin in wt mice results in a significant supra-functional benefit over wt. These findings highlight an additive benefit of the combined therapy and potential new unique roles of utrophin. Finally, we show a 30% restoration of wt dystrophin levels, using exon-skipping, together with increased utrophin levels restores dystrophic muscle function to wt levels offering greater therapeutic benefit than either single approach alone. Thus, this combination therapy results in additive functional benefit and paves the way for potential future combinations of dystrophin- and utrophin-based strategies.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/therapy , Utrophin/genetics , Animals , Dystrophin/metabolism , Exons , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Morpholinos/chemical synthesis , Morpholinos/therapeutic use , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myofibrils/metabolism , Sarcolemma/metabolism , Up-Regulation , Utrophin/metabolism
7.
Hum Mol Genet ; 28(2): 307-319, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30304405

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. Constitutive utrophin expression, a structural and functional paralogue of dystrophin, can successfully prevent the dystrophic pathology in the dystrophin-deficient mdx mouse model. In dystrophic muscles, utrophin is increased as part of the repair process and localized at the sarcolemma of regenerating myofibers. The presence of developmental myosin such as embryonic myosin (MyHC-emb) and neonatal represents a useful marker of muscle regeneration and a meaningful indicator of muscle damage, which correlates with the clinical severity of milder Becker muscular dystrophy and DMD patients. In the present study, we demonstrate that MyHC-emb is a robust marker of regeneration at different ages and in different skeletal muscles. We also evaluate the correlation between utrophin, dystrophin and MyHC-emb in wild-type (wt) and regenerating dystrophic muscles. Restoration of dystrophin significantly reduced MyHC-emb levels. Similarly, overexpression of utrophin in the transgenic mdx-Fiona mice reduced the number of MyHC-emb positive fibers to wt level, prevented the regenerative process and rescued the muscle function. In contrast, the absence of utrophin in the dystrophin-deficient double-knockout mice resulted in a higher MyHC-emb content and in a more severe dystrophic pathophysiology than in mdx mice. These data illustrate the importance of monitoring utrophin and MyHC-emb levels in the preclinical evaluation of therapies and provide translational support for the use of developmental myosin as a disease biomarker in DMD clinical trials.


Subject(s)
Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myosins/metabolism , Regeneration , Utrophin/metabolism , Animals , Biomarkers/metabolism , Clinical Trials as Topic , Disease Models, Animal , Embryo, Mammalian/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/embryology , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/metabolism
8.
Mol Ther Methods Clin Dev ; 11: 92-105, 2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30417024

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by mutations in the dystrophin gene. DMD boys are wheelchair-bound around 12 years and generally survive into their twenties. There is currently no effective treatment except palliative care, although personalized treatments such as exon skipping, stop codon read-through, and viral-based gene therapies are making progress. Patients present with skeletal muscle pathology, but most also show cardiomyopathy by the age of 10. A systemic therapeutic approach is needed that treats the heart and skeletal muscle defects in all patients. The dystrophin-related protein utrophin has been shown to compensate for the lack of dystrophin in the mildly affected BL10/mdx mouse. The purpose of this investigation was to demonstrate that AAV9-mediated micro-utrophin transgene delivery can not only functionally replace dystrophin in the heart, but also attenuate the skeletal muscle phenotype in severely affected D2/mdx mice. The data presented here show that utrophin can indeed alleviate the pathology in skeletal and cardiac muscle in D2/mdx mice. These results endorse the view that utrophin modulation has the potential to increase the quality life of all DMD patients whatever their mutation.

9.
Hum Mol Genet ; 27(10): 1723-1731, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29509900

ABSTRACT

Polyglutamine expansions in the huntingtin gene cause Huntington's disease (HD). Huntingtin is ubiquitously expressed, leading to pathological alterations also in peripheral organs. Variations in the length of the polyglutamine tract explain up to 70% of the age-at-onset variance, with the rest of the variance attributed to genetic and environmental modifiers. To identify novel disease modifiers, we performed an unbiased mutagenesis screen on an HD mouse model, identifying a mutation in the skeletal muscle voltage-gated sodium channel (Scn4a, termed 'draggen' mutation) as a novel disease enhancer. Double mutant mice (HD; Scn4aDgn/+) had decreased survival, weight loss and muscle atrophy. Expression patterns show that the main tissue affected is skeletal muscle. Intriguingly, muscles from HD; Scn4aDgn/+ mice showed adaptive changes similar to those found in endurance exercise, including AMPK activation, fibre type switching and upregulation of mitochondrial biogenesis. Therefore, we evaluated the effects of endurance training on HD mice. Crucially, this training regime also led to detrimental effects on HD mice. Overall, these results reveal a novel role for skeletal muscle in modulating systemic HD pathogenesis, suggesting that some forms of physical exercise could be deleterious in neurodegeneration.


Subject(s)
Huntington Disease/genetics , Muscular Atrophy/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Animals , Disease Models, Animal , Endurance Training , Enhancer Elements, Genetic , Humans , Huntingtin Protein/genetics , Huntington Disease/physiopathology , Huntington Disease/therapy , Mice , Muscular Atrophy/physiopathology , Muscular Atrophy/therapy , Mutation , Neurons/pathology , Neurons/physiology , Organelle Biogenesis , Peptides/genetics , Physical Conditioning, Animal , Trinucleotide Repeat Expansion/genetics
10.
Mol Metab ; 6(11): 1419-1428, 2017 11.
Article in English | MEDLINE | ID: mdl-29107289

ABSTRACT

OBJECTIVE: Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. METHODS: In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. RESULTS: We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. CONCLUSIONS: OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Anxiety/metabolism , Base Sequence , Brain/metabolism , Chromosome Mapping , Databases, Genetic , Female , Gene Expression , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox , Homeodomain Proteins/physiology , Humans , Hypothalamus/metabolism , Male , Mice , Nerve Tissue Proteins/physiology , Neurosecretory Systems/metabolism , Obesity/metabolism , Transcription Factors/genetics , Transcriptome/genetics
11.
Skelet Muscle ; 7(1): 22, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29065908

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a lethal X-linked muscle wasting disorder caused by the absence of dystrophin, a large cytoskeletal muscle protein. Increasing the levels of the dystrophin-related-protein utrophin is a highly promising therapy for DMD and has been shown to improve pathology in dystrophin-deficient mice. One contributing factor to muscle wasting in DMD is mitochondrial pathology that contributes to oxidative stress and propagates muscle damage. The purpose of this study was to assess whether utrophin could attenuate mitochondria pathology and oxidative stress. METHODS: Skeletal muscles from wildtype C57BL/10, dystrophin-deficient mdx, dystrophin/utrophin double knockout (dko) and dystrophin-deficient mdx/utrophin over-expressing mdx-Fiona transgenic mice were assessed for markers of mitochondrial damage. RESULTS: Using transmission electron microscopy, we show that high levels of utrophin ameliorate the aberrant structure and localisation of mitochondria in mdx mice whereas absence of utrophin worsened these features in dko mice. Elevated utrophin also reverts markers of protein oxidation and oxidative stress, elevated in mdx and dko mice, to wildtype levels. These changes were observed independently of a shift in oxidative phenotype. CONCLUSION: These findings show that utrophin levels influence mitochondrial pathology and oxidative stress. While utrophin deficiency worsens the pathology, utrophin over-expression in dystrophic muscle benefits mitochondria and attenuates the downstream pathology associated with aberrant mitochondrial function.


Subject(s)
Mitochondria, Muscle/metabolism , Muscular Dystrophy, Duchenne/metabolism , Oxidative Stress , Utrophin/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria, Muscle/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Utrophin/metabolism
12.
Soft Matter ; 13(40): 7441-7452, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-28967661

ABSTRACT

A small library of thermoresponsive amphiphilic copolymers based on polylactide-block-poly((2-(2-methoxyethoxy)ethyl methacrylate)-co-(oligoethylene glycol methacrylate)) (PLA-b-P(DEGMA)-co-(OEGMA)), was synthesised by copper-mediated controlled radical polymerisation (CRP) with increasing ratios of OEGMA : DEGMA. These polymers were combined in two ways to form nanoparticles with controllable thermal transition temperatures as measured by particle aggregation. The first technique involved the blending of two (PLA-b-P(DEGMA)-co-(OEGMA)) polymers together prior to assembling nanoparticles (NPs). The second method involved mixing pre-formed nanoparticles of single (PLA-b-P(DEGMA)-co-(OEGMA)) polymers. The observed critical aggregation temperature Tt did not change in a linear relationship with the ratios of each copolymer either in the nanoparticles blended from different copolymers or in the mixtures of pre-formed nanoparticles. However, where co-polymer mixtures were based on (OEG)9MA ratios within 5-10 mole%, a linear relationship between (OEG)9MA composition in the blends and Tt was obtained. The data suggest that OEGMA-based copolymers are tunable over a wide temperature range given suitable co-monomer content in the linear polymers or nanoparticles. Moreover, the thermal transitions of the nanoparticles were reversible and repeatable, with the cloud point curves being essentially invariant across at least three heating and cooling cycles, and a selected nanoparticle formulation was found to be readily endocytosed in representative cancer cells and fibroblasts.


Subject(s)
Biocompatible Materials/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Temperature , Biocompatible Materials/toxicity , Humans , MCF-7 Cells , Polymers/toxicity
13.
Curr Protoc Mouse Biol ; 6(3): 211-222, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27584551

ABSTRACT

In mice with altered body composition, establishing whether it is food intake or energy expenditure, or both, that is the major determinant resulting in changed energy balance is important. In order to ascertain where the imbalance is, the acquisition of reproducible data is critical. Therefore, here we provide detailed descriptions of how to determine energy balance in mice. This encompasses protocols for establishing energy intake from home cage measurement of food intake, determining energy lost in feces using bomb calorimetry, and using equations to calculate parameters such as energy intake (EI), digested energy intake (DEI), and metabolisable energy intake (MEI) to determine overall energy balance. We also discuss considerations that should be taken into account when planning these experiments, including diet and sample sizes. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Eating , Energy Intake , Energy Metabolism , Mice/physiology , Physiology/methods , Animals , Body Composition , Calorimetry , Feces/chemistry , Models, Animal , Models, Biological
14.
PLoS One ; 8(6): e61870, 2013.
Article in English | MEDLINE | ID: mdl-23826075

ABSTRACT

We employed a random mutagenesis approach to identify novel monogenic determinants of type 2 diabetes. Here we show that haplo-insufficiency of the histone methyltransferase myeloid-lineage leukemia (Mll2/Wbp7) gene causes type 2 diabetes in the mouse. We have shown that mice heterozygous for two separate mutations in the SET domain of Mll2 or heterozygous Mll2 knockout mice were hyperglycaemic, hyperinsulinaemic and developed non-alcoholic fatty liver disease. Consistent with previous Mll2 knockout studies, mice homozygous for either ENU mutation (or compound heterozygotes) died during embryonic development at 9.5-14.5 days post coitum. Heterozygous deletion of Mll2 induced in the adult mouse results in a normal phenotype suggesting that changes in chromatin methylation during development result in the adult phenotype. Mll2 has been shown to regulate a small subset of genes, a number of which Neurod1, Enpp1, Slc27a2, and Plcxd1 are downregulated in adult mutant mice. Our results demonstrate that histone H3K4 methyltransferase Mll2 is a component of the genetic regulation necessary for glucose homeostasis, resulting in a specific disease pattern linking chromatin modification with causes and progression of type 2 diabetes, providing a basis for its further understanding at the molecular level.


Subject(s)
Glucose Tolerance Test , Insulin Resistance/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Amino Acid Sequence , Animals , Genes, Lethal , Histone-Lysine N-Methyltransferase , Islets of Langerhans/pathology , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Myeloid-Lymphoid Leukemia Protein/chemistry , Polymerase Chain Reaction , Sequence Homology, Amino Acid
15.
Curr Diab Rep ; 12(6): 651-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22996130

ABSTRACT

The genomes of many species have now been completely sequenced including human and mouse. Great progress has been made in understanding the complex genetics that underlie diabetes and obesity in human populations. One of the current challenges is the functional identification and characterization of the genes within loci that are being mapped. There are many approaches to this problem and this review outlines the valuable role that the mouse can play. We outline the mouse resources that are available to the research community, including knockouts with conditional potential for every gene, and the efforts of the International Mouse Phenotyping Consortium to attach phenotype information to these genes. We also briefly consider the potential of TALEN technology to tailor-make new mouse models of specific mutations discovered in humans. Finally, we consider the recent progress in characterizing the GWAS genes FTO, TCF7L2, CDKAL1, and SLC30A8 in engineered mouse models.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Genome-Wide Association Study , Obesity/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Cyclin-Dependent Kinase 5/genetics , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , Mixed Function Oxygenases/genetics , Nerve Tissue Proteins/genetics , Oxo-Acid-Lyases/genetics , Proteins/genetics , Transcription Factor 7-Like 2 Protein/genetics , tRNA Methyltransferases
16.
Mol Cell Biol ; 32(5): 1017-29, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22215617

ABSTRACT

The imprinted Gnas cluster is involved in obesity, energy metabolism, feeding behavior, and viability. Relative contribution of paternally expressed proteins XLαs, XLN1, and ALEX or a double dose of maternally expressed Gsα to phenotype has not been established. In this study, we have generated two new mutants (Ex1A-T-CON and Ex1A-T) at the Gnas cluster. Paternal inheritance of Ex1A-T-CON leads to loss of imprinting of Gsα, resulting in preweaning growth retardation followed by catch-up growth. Paternal inheritance of Ex1A-T leads to loss of imprinting of Gsα and loss of expression of XLαs and XLN1. These mice have severe preweaning growth retardation and incomplete catch-up growth. They are fully viable probably because suckling is unimpaired, unlike mutants in which the expression of all the known paternally expressed Gnasxl proteins (XLαs, XLN1 and ALEX) is compromised. We suggest that loss of ALEX is most likely responsible for the suckling defects previously observed. In adults, paternal inheritance of Ex1A-T results in an increased metabolic rate and reductions in fat mass, leptin, and bone mineral density attributable to loss of XLαs. This is, to our knowledge, the first report describing a role for XLαs in bone metabolism. We propose that XLαs is involved in the regulation of bone and adipocyte metabolism.


Subject(s)
Energy Metabolism/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Lipid Metabolism/genetics , Obesity/genetics , Animals , Bone and Bones/metabolism , Chromogranins , Female , Gene Dosage , Genetic Loci , Genomic Imprinting , Male , Mice , Mice, Knockout , Protein Isoforms/genetics , Sucking Behavior
17.
Nat Genet ; 42(12): 1086-92, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21076408

ABSTRACT

Genome-wide association studies have identified SNPs within FTO, the human fat mass and obesity-associated gene, that are strongly associated with obesity. Individuals homozygous for the at-risk rs9939609 A allele weigh, on average, ~3 kg more than individuals with the low-risk T allele. Mice that lack FTO function and/or Fto expression display increased energy expenditure and a lean phenotype. We show here that ubiquitous overexpression of Fto leads to a dose-dependent increase in body and fat mass, irrespective of whether mice are fed a standard or a high-fat diet. Our results suggest that increased body mass results primarily from increased food intake. Mice with increased Fto expression on a high-fat diet develop glucose intolerance. This study provides the first direct evidence that increased Fto expression causes obesity in mice.


Subject(s)
Feeding Behavior/physiology , Obesity/genetics , Oxo-Acid-Lyases/metabolism , Adiposity/drug effects , Adiposity/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Area Under Curve , Body Temperature , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Feeding Behavior/drug effects , Female , Glucose/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Male , Mice , Mixed Function Oxygenases , Models, Animal , Motor Activity/drug effects , Obesity/blood , Oxo-Acid-Lyases/genetics
18.
PLoS Genet ; 5(8): e1000599, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19680540

ABSTRACT

Human FTO gene variants are associated with body mass index and type 2 diabetes. Because the obesity-associated SNPs are intronic, it is unclear whether changes in FTO expression or splicing are the cause of obesity or if regulatory elements within intron 1 influence upstream or downstream genes. We tested the idea that FTO itself is involved in obesity. We show that a dominant point mutation in the mouse Fto gene results in reduced fat mass, increased energy expenditure, and unchanged physical activity. Exposure to a high-fat diet enhances lean mass and lowers fat mass relative to control mice. Biochemical studies suggest the mutation occurs in a structurally novel domain and modifies FTO function, possibly by altering its dimerisation state. Gene expression profiling revealed increased expression of some fat and carbohydrate metabolism genes and an improved inflammatory profile in white adipose tissue of mutant mice. These data provide direct functional evidence that FTO is a causal gene underlying obesity. Compared to the reported mouse FTO knockout, our model more accurately reflects the effect of human FTO variants; we observe a heterozygous as well as homozygous phenotype, a smaller difference in weight and adiposity, and our mice do not show perinatal lethality or an age-related reduction in size and length. Our model suggests that a search for human coding mutations in FTO may be informative and that inhibition of FTO activity is a possible target for the treatment of morbid obesity.


Subject(s)
Adipose Tissue/metabolism , Obesity/genetics , Obesity/metabolism , Oxo-Acid-Lyases/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Amino Acid Sequence , Animals , Body Weight , Disease Models, Animal , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mixed Function Oxygenases , Molecular Sequence Data , Mutation, Missense , Obesity/physiopathology , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Protein Structure, Tertiary , Sequence Alignment
19.
Diabetes ; 53(6): 1577-83, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161764

ABSTRACT

Here we report the first cloned N-ethyl-nitrosourea (ENU)-derived mouse model of diabetes. GENA348 was identified through free-fed plasma glucose measurement, being more than 2 SDs above the population mean of a cohort of >1,201 male ENU mutant mice. The underlying gene was mapped to the maturity-onset diabetes of the young (MODY2) homology region of mouse chromosome 11 (logarithm of odds 6.0). Positional candidate gene analyses revealed an A to T transversion mutation in exon 9 of the glucokinase gene, resulting in an isoleucine to phenylalanine change at amino acid 366 (I366F). Heterozygous mutants have 67% of the enzyme activity of wild-type littermates (P < 0.0012). Homozygous mutants have less enzyme activity (14% of wild-type activity) and are even less glucose tolerant. The GENA348 allele is novel because no mouse or human diabetes studies have described a mutation in the corresponding amino acid position. It is also the first glucokinase missense mutation reported in mice and is homozygous viable, unlike the global knockout mutations. This work demonstrates that ENU mutagenesis screens can be used to generate models of complex phenotypes, such as type 2 diabetes, that are directly relevant to human disease.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Ethylnitrosourea/pharmacology , Glucokinase/genetics , Mutagens/pharmacology , Mutation, Missense , Adenine , Amino Acid Sequence , Amino Acid Substitution , Animals , Chromosome Mapping , Glucokinase/drug effects , Glucokinase/metabolism , Glucose/metabolism , Glucose Intolerance/genetics , Heterozygote , Homozygote , Isoleucine , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Phenylalanine , Phosphorylation , Thymine
20.
Nat Genet ; 34(4): 421-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12833159

ABSTRACT

The whirler mouse mutant (wi) does not respond to sound stimuli, and detailed ultrastructural analysis of sensory hair cells in the organ of Corti of the inner ear indicates that the whirler gene encodes a protein involved in the elongation and maintenance of stereocilia in both inner hair cells (IHCs) and outer hair cells (OHCs). BAC-mediated transgene correction of the mouse phenotype and mutation analysis identified the causative gene as encoding a novel PDZ protein called whirlin. The gene encoding whirlin also underlies the human autosomal recessive deafness locus DFNB31. In the mouse cochlea, whirlin is expressed in the sensory IHC and OHC stereocilia. Our findings suggest that this novel PDZ domain-containing molecule acts as an organizer of submembranous molecular complexes that control the coordinated actin polymerization and membrane growth of stereocilia.


Subject(s)
Deafness/genetics , Gene Expression , Membrane Proteins/genetics , Proteins/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cilia/physiology , Cilia/ultrastructure , DNA Mutational Analysis , DNA, Complementary/genetics , Genes, Recessive , Hair Cells, Auditory, Inner/ultrastructure , Hair Cells, Auditory, Outer/ultrastructure , Humans , Membrane Proteins/physiology , Mice , Mice, Mutant Strains , Mice, Transgenic , Molecular Sequence Data , Phenotype , Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...