Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38915588

ABSTRACT

ECHS1 Deficiency (ECHS1D) is a rare and devastating pediatric disease that currently has no defined treatments. This disorder results from missense loss-of-function mutations in the ECHS1 gene that result in severe developmental delays, encephalopathy, hypotonia, and early death. ECHS1 enzymatic activity is necessary for the beta-oxidation of fatty acids and the oxidation of branched-chain amino acids within the inner mitochondrial matrix. The pathogenesis of disease remains unknown, however it is hypothesized that disease is driven by an accumulation of toxic metabolites from impaired valine oxidation. To expand our knowledge on disease mechanisms, a novel mouse model of ECHS1D was generated that possesses a disease-associated knock-in (KI) allele and a knock-out (KO) allele. To investigate the behavioral phenotype, a battery of testing was performed at multiple time points, which included assessments of learning, motor function, endurance, sensory responses, and anxiety. Neurological abnormalities were assessed using wireless telemetry EEG recordings, pentylenetetrazol (PTZ) seizure induction, and immunohistochemistry. Metabolic perturbations were measured within the liver, serum, and brain using mass spectrometry and magnetic resonance spectroscopy. To test disease mechanisms, mice were subjected to disease pathway stressors and then survival, body weight gain, and epilepsy were assessed. Mice containing KI/KI or KI/KO alleles were viable with normal development and survival, and the presence of KI and KO alleles resulted in a significant reduction in ECHS1 protein. ECHS1D mice displayed reduced exercise capacity and pain sensation. EEG analysis revealed increased slow wave power that was associated with perturbations in sleep. ECHS1D mice had significantly increased epileptiform EEG discharges, and were sensitive to seizure induction, which resulted in death of 60% of ECHS1D mice. Under basal conditions, brain structure was grossly normal, although histological analysis revealed increased microglial activation in aged ECHS1D mice. Increased dietary valine only affected ECHS1D mice, which significantly exacerbated seizure susceptibility and resulted in death. Lastly, acute inflammatory challenge drove regression and early lethality in ECHS1D mice. In conclusion, we developed a novel model of ECHS1D that may be used to further knowledge on disease mechanisms and to develop therapeutics. Our data suggests altered metabolic signaling and inflammation may contribute to epilepsy in ECHS1D, and these alterations may be attributed to impaired valine metabolism.

2.
Sci Transl Med ; 16(748): eadk1358, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776392

ABSTRACT

Blood-CNS barrier disruption is a hallmark of numerous neurological disorders, yet whether barrier breakdown is sufficient to trigger neurodegenerative disease remains unresolved. Therapeutic strategies to mitigate barrier hyperpermeability are also limited. Dominant missense mutations of the cation channel transient receptor potential vanilloid 4 (TRPV4) cause forms of hereditary motor neuron disease. To gain insights into the cellular basis of these disorders, we generated knock-in mouse models of TRPV4 channelopathy by introducing two disease-causing mutations (R269C and R232C) into the endogenous mouse Trpv4 gene. TRPV4 mutant mice exhibited weakness, early lethality, and regional motor neuron loss. Genetic deletion of the mutant Trpv4 allele from endothelial cells (but not neurons, glia, or muscle) rescued these phenotypes. Symptomatic mutant mice exhibited focal disruptions of blood-spinal cord barrier (BSCB) integrity, associated with a gain of function of mutant TRPV4 channel activity in neural vascular endothelial cells (NVECs) and alterations of NVEC tight junction structure. Systemic administration of a TRPV4-specific antagonist abrogated channel-mediated BSCB impairments and provided a marked phenotypic rescue of symptomatic mutant mice. Together, our findings show that mutant TRPV4 channels can drive motor neuron degeneration in a non-cell autonomous manner by precipitating focal breakdown of the BSCB. Further, these data highlight the reversibility of TRPV4-mediated BSCB impairments and identify a potential therapeutic strategy for patients with TRPV4 mutations.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Gain of Function Mutation , Motor Neurons , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Motor Neurons/pathology , Motor Neurons/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Mice , Nerve Degeneration/pathology , Nerve Degeneration/genetics , Phenotype , Spinal Cord/pathology , Spinal Cord/metabolism
3.
bioRxiv ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38260608

ABSTRACT

KCNQ2 variants in children with neurodevelopmental impairment are difficult to assess due to their heterogeneity and unclear pathogenic mechanisms. We describe a child with neonatal-onset epilepsy, developmental impairment of intermediate severity, and KCNQ2 G256W heterozygosity. Analyzing prior KCNQ2 channel cryoelectron microscopy models revealed G256 as a node of an arch-shaped non-covalent bond network linking S5, the pore turret, and the ion path. Co-expression with G256W dominantly suppressed conduction by wild-type subunits in heterologous cells. Ezogabine partly reversed this suppression. G256W/+ mice have epilepsy leading to premature deaths. Hippocampal CA1 pyramidal cells from G256W/+ brain slices showed hyperexcitability. G256W/+ pyramidal cell KCNQ2 and KCNQ3 immunolabeling was significantly shifted from axon initial segments to neuronal somata. Despite normal mRNA levels, G256W/+ mouse KCNQ2 protein levels were reduced by about 50%. Our findings indicate that G256W pathogenicity results from multiplicative effects, including reductions in intrinsic conduction, subcellular targeting, and protein stability. These studies provide evidence for an unexpected and novel role for the KCNQ2 pore turret and introduce a valid animal model of KCNQ2 encephalopathy. Our results, spanning structure to behavior, may be broadly applicable because the majority of KCNQ2 encephalopathy patients share variants near the selectivity filter.

4.
Neurobiol Dis ; 177: 105996, 2023 02.
Article in English | MEDLINE | ID: mdl-36638893

ABSTRACT

Friedreich's ataxia (FRDA) is a neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin (FXN). Most FRDA patients are homozygous for large expansions of GAA repeats in intron 1 of FXN, while some are compound heterozygotes with an expanded GAA tract in one allele and a missense or nonsense mutation in the other. A missense mutation, changing a glycine to valine at position 130 (G130V), is prevalent among the clinical variants. We and others have demonstrated that levels of mature FXN protein in FRDA G130V samples are reduced below those detected in samples harboring homozygous repeat expansions. Little is known regarding expression and function of endogenous FXN-G130V protein due to lack of reagents and models that can distinguish the mutant FXN protein from the wild-type FXN produced from the GAA-expanded allele. We aimed to determine the effect of the G130V (murine G127V) mutation on Fxn expression and to define its multi-system impact in vivo. We used CRISPR/Cas9 to introduce the G127V missense mutation in the Fxn coding sequence and generated homozygous mice (FxnG127V/G127V). We also introduced the G127V mutation into a GAA repeat expansion FRDA mouse model (FxnGAA230/KO; KIKO) to generate a compound heterozygous strain (FxnG127V/GAA230). We performed neurobehavioral tests on cohorts of WT and Fxn mutant animals at three-month intervals for one year, and collected tissue samples to analyze molecular changes during that time. The endogenous Fxn G127V protein is detected at much lower levels in all tissues analyzed from FxnG127V/G127V mice compared to age and sex-matched WT mice without differences in Fxn transcript levels. FxnG127V/G127V mice are significantly smaller than WT counterparts, but perform similarly in most neurobehavioral tasks. RNA sequencing analysis revealed reduced expression of genes in oxidative phosphorylation and protein synthesis, underscoring the metabolic consequences in our mouse model expressing extremely low levels of Fxn. Results of these studies provide insight into the unique pathogenic mechanism of the FXN G130V mechanism and the tolerable limit of Fxn/FXN expression in vivo.


Subject(s)
Friedreich Ataxia , Neurodegenerative Diseases , Mice , Animals , Neurodegenerative Diseases/genetics , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Protein Biosynthesis , Disease Models, Animal , Friedreich Ataxia/metabolism , Trinucleotide Repeat Expansion , Frataxin
5.
Biochem Biophys Res Commun ; 645: 164-172, 2023 02 19.
Article in English | MEDLINE | ID: mdl-36689813

ABSTRACT

Matrin 3 is a nuclear matrix protein that has many roles in RNA processing including splicing and transport of mRNA. Many missense mutations in the Matrin 3 gene (MATR3) have been linked to familial forms of amyotrophic lateral sclerosis (ALS) and distal myopathy. However, the exact role of MATR3 mutations in ALS and myopathy pathogenesis is not understood. To demonstrate a role of MATR3 mutations in vivo, we generated a novel CRISPR/Cas9 mediated knock-in mouse model harboring the MATR3 P154S mutation expressed under the control of the endogenous promoter. The P154S variant of the MATR3 gene has been linked to familial forms of ALS. Heterozygous and homozygous MATR3 P154S knock-in mice did not develop progressive motor deficits compared to wild-type mice. In addition, ALS-like pathology did not develop in nervous or muscle tissue in either heterozygous or homozygous mice. Our results suggest that the MATR3 P154S variant is not sufficient to produce ALS-like pathology in vivo.


Subject(s)
Amyotrophic Lateral Sclerosis , Nuclear Matrix-Associated Proteins , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , Muscles/metabolism , Muscular Diseases/genetics , Mutation , Mutation, Missense , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism
6.
bioRxiv ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168388

ABSTRACT

Loss of nuclear TDP-43 occurs in a wide range of neurodegenerative diseases, and specific mutations in the TARDBP gene that encodes the protein are linked to familial Frontal Temporal Lobar Dementia (FTD), and Amyotrophic Lateral Sclerosis (ALS). Although the focus has been on neuronal cell dysfunction caused by TDP-43 variants, TARDBP mRNA transcripts are expressed at similar levels in brain endothelial cells (ECs). Since increased permeability across the blood brain barrier (BBB) precedes cognitive decline, we postulated that altered functions of TDP-43 in ECs contributes to BBB dysfunction in neurodegenerative disease. To test this hypothesis, we examined EC function and BBB properties in mice with either knock-in mutations found in ALS/FTLD patients (TARDBPG348C and GRNR493X) or EC-specific deletion of TDP-43 throughout the endothelium (Cdh5(PAC)CreERT2; Tardbpff) or restricted to brain endothelium (Slco1c1(BAC)CreERT2; Tardbpff). We found that TARDBPG348C mice exhibited increased permeability to 3kDa Texas Red dextran and NHS-biotin, relative to their littermate controls, which could be recapitulated in cultured brain ECs from these mice. Nuclear levels of TDP-43 were reduced in vitro and in vivo in ECs from TARDBPG348C mice. This coincided with a reduction in junctional proteins VE-cadherin, claudin-5 and ZO-1 in isolated ECs, supporting a cell autonomous effect on barrier function through a loss of nuclear TDP-43. We further examined two models of Tardbp deletion in ECs, and found that the loss of TDP-43 throughout the endothelium led to systemic endothelial activation and permeability. Deletion specifically within the brain endothelium acutely increased BBB permeability, and eventually led to hallmarks of FTD, including fibrin deposition, microglial and astrocyte activation, and behavioral defects. Together, these data show that TDP-43 dysfunction specifically within brain ECs would contribute to the BBB defects observed early in the progression of ALS/FTLD.

7.
Elife ; 92020 12 14.
Article in English | MEDLINE | ID: mdl-33315011

ABSTRACT

N-Glycanase 1 (NGLY1) is a cytoplasmic deglycosylating enzyme. Loss-of-function mutations in the NGLY1 gene cause NGLY1 deficiency, which is characterized by developmental delay, seizures, and a lack of sweat and tears. To model the phenotypic variability observed among patients, we crossed a Drosophila model of NGLY1 deficiency onto a panel of genetically diverse strains. The resulting progeny showed a phenotypic spectrum from 0 to 100% lethality. Association analysis on the lethality phenotype, as well as an evolutionary rate covariation analysis, generated lists of modifying genes, providing insight into NGLY1 function and disease. The top association hit was Ncc69 (human NKCC1/2), a conserved ion transporter. Analyses in NGLY1-/- mouse cells demonstrated that NKCC1 has an altered average molecular weight and reduced function. The misregulation of this ion transporter may explain the observed defects in secretory epithelium function in NGLY1 deficiency patients.


Subject(s)
Congenital Disorders of Glycosylation/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Solute Carrier Family 12, Member 2/metabolism , Animals , Disease Models, Animal , Drosophila melanogaster , Mice , Mice, Knockout , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Phenotype
8.
EMBO Rep ; 21(10): e50197, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32761777

ABSTRACT

Progranulin (PGRN) and transmembrane protein 106B (TMEM106B) are important lysosomal proteins implicated in frontotemporal lobar degeneration (FTLD) and other neurodegenerative disorders. Loss-of-function mutations in progranulin (GRN) are a common cause of FTLD, while TMEM106B variants have been shown to act as disease modifiers in FTLD. Overexpression of TMEM106B leads to lysosomal dysfunction, while loss of Tmem106b ameliorates lysosomal and FTLD-related pathologies in young Grn-/- mice, suggesting that lowering TMEM106B might be an attractive strategy for therapeutic treatment of FTLD-GRN. Here, we generate and characterize older Tmem106b-/- Grn-/- double knockout mice, which unexpectedly show severe motor deficits and spinal cord motor neuron and myelin loss, leading to paralysis and premature death at 11-12 months. Compared to Grn-/- , Tmem106b-/- Grn-/- mice have exacerbated FTLD-related pathologies, including microgliosis, astrogliosis, ubiquitin, and phospho-Tdp43 inclusions, as well as worsening of lysosomal and autophagic deficits. Our findings confirm a functional interaction between Tmem106b and Pgrn and underscore the need to rethink whether modulating TMEM106B levels is a viable therapeutic strategy.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Animals , Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins , Progranulins/genetics
9.
Dis Model Mech ; 13(7)2020 07 27.
Article in English | MEDLINE | ID: mdl-32586831

ABSTRACT

Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin (FXN). Most FRDA patients are homozygous for large expansions of GAA repeat sequences in intron 1 of FXN, whereas a fraction of patients are compound heterozygotes, with a missense or nonsense mutation in one FXN allele and expanded GAAs in the other. A prevalent missense mutation among FRDA patients changes a glycine at position 130 to valine (G130V). Herein, we report generation of the first mouse model harboring an Fxn point mutation. Changing the evolutionarily conserved glycine 127 in mouse Fxn to valine results in a failure-to-thrive phenotype in homozygous animals and a substantially reduced number of offspring. Like G130V in FRDA, the G127V mutation results in a dramatic decrease of Fxn protein without affecting transcript synthesis or splicing. FxnG127V mouse embryonic fibroblasts exhibit significantly reduced proliferation and increased cell senescence. These defects are evident in early passage cells and are exacerbated at later passages. Furthermore, increased frequency of mitochondrial DNA lesions and fragmentation are accompanied by marked amplification of mitochondrial DNA in FxnG127V cells. Bioenergetics analyses demonstrate higher sensitivity and reduced cellular respiration of FxnG127V cells upon alteration of fatty acid availability. Importantly, substitution of FxnWT with FxnG127V is compatible with life, and cellular proliferation defects can be rescued by mitigation of oxidative stress via hypoxia or induction of the NRF2 pathway. We propose FxnG127V cells as a simple and robust model for testing therapeutic approaches for FRDA.


Subject(s)
Cell Proliferation , Cellular Senescence , Fibroblasts/pathology , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Mitochondria/pathology , Point Mutation , Animals , Cell Line , Disease Models, Animal , Energy Metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Genetic Predisposition to Disease , Iron-Binding Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phenotype , Frataxin
10.
J Clin Invest ; 130(3): 1506-1512, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32065591

ABSTRACT

Notch signaling is a highly conserved intercellular pathway with tightly regulated and pleiotropic roles in normal tissue development and homeostasis. Dysregulated Notch signaling has also been implicated in human disease, including multiple forms of cancer, and represents an emerging therapeutic target. Successful development of such therapeutics requires a detailed understanding of potential on-target toxicities. Here, we identify autosomal dominant mutations of the canonical Notch ligand Jagged1 (or JAG1) as a cause of peripheral nerve disease in 2 unrelated families with the hereditary axonal neuropathy Charcot-Marie-Tooth disease type 2 (CMT2). Affected individuals in both families exhibited severe vocal fold paresis, a rare feature of peripheral nerve disease that can be life-threatening. Our studies of mutant protein posttranslational modification and localization indicated that the mutations (p.Ser577Arg, p.Ser650Pro) impair protein glycosylation and reduce JAG1 cell surface expression. Mice harboring heterozygous CMT2-associated mutations exhibited mild peripheral neuropathy, and homozygous expression resulted in embryonic lethality by midgestation. Together, our findings highlight a critical role for JAG1 in maintaining peripheral nerve integrity, particularly in the recurrent laryngeal nerve, and provide a basis for the evaluation of peripheral neuropathy as part of the clinical development of Notch pathway-modulating therapeutics.


Subject(s)
Charcot-Marie-Tooth Disease , Genes, Dominant , Jagged-1 Protein , Mutation, Missense , Signal Transduction/genetics , Amino Acid Substitution , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Female , Glycosylation , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Mice , Receptors, Notch/genetics , Receptors, Notch/metabolism
11.
Proc Natl Acad Sci U S A ; 108(44): 18108-13, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22025706

ABSTRACT

The anorectic anx/anx mouse exhibits disturbed feeding behavior and aberrances, including neurodegeneration, in peptidergic neurons in the appetite regulating hypothalamic arcuate nucleus. Poor feeding in infants, as well as neurodegeneration, are common phenotypes in human disorders caused by dysfunction of the mitochondrial oxidative phosphorylation system (OXPHOS). We therefore hypothesized that the anorexia and degenerative phenotypes in the anx/anx mouse could be related to defects in the OXPHOS. In this study, we found reduced efficiency of hypothalamic OXPHOS complex I assembly and activity in the anx/anx mouse. We also recorded signs of increased oxidative stress in anx/anx hypothalamus, possibly as an effect of the decreased hypothalamic levels of fully assembled complex I, that were demonstrated by native Western blots. Furthermore, the Ndufaf1 gene, encoding a complex I assembly factor, was genetically mapped to the anx interval and found to be down-regulated in anx/anx mice. These results suggest that the anorexia and hypothalamic neurodegeneration of the anx/anx mouse are associated with dysfunction of mitochondrial complex I.


Subject(s)
Anorexia/physiopathology , Hypothalamus/physiopathology , Mitochondria/physiology , Alleles , Animals , Anorexia/genetics , Hypothalamus/metabolism , Mice , Mitochondria/metabolism , Oxidative Phosphorylation , Oxidative Stress
12.
Mamm Genome ; 21(1-2): 52-63, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20033694

ABSTRACT

C57BL/6 J (B6) and CAST/EiJ (CAST), the inbred strain derived from M. musculus castaneus, differ in nutrient intake behaviors, including dietary fat and carbohydrate consumption in a two-diet-choice paradigm. Significant quantitative trait loci (QTLs) for carbohydrate (Mnic1) and total energy intake (Kcal2) are present between these strains on chromosome (Chr) 17. Here we report the refinement of the Chr 17 QTL in a subcongenic strain of the B6.CAST-( D17Mit19-D17Mit91 ) congenic mice described previously. This new subcongenic strain possesses CAST Chr 17 donor alleles from 4.8 to 45.4 Mb on a B6 background. Similar to CAST, the subcongenic mice exhibit increased carbohydrate and total calorie intake per body weight, while fat intake remains equivalent. Unexpectedly, this CAST genomic segment also confers two new physical activity phenotypes: 22% higher spontaneous physical activity levels and significantly increased voluntary wheel-running activity compared with the parental B6 strain. Overall, these data suggest that gene(s) involved in carbohydrate preference and increased physical activity are contained within the proximal region of Chr 17. Interval-specific microarray analysis in hypothalamus and skeletal muscle revealed differentially expressed genes within the subcongenic region, including neuropeptide W (Npw); glyoxalase I (Glo1); cytochrome P450, family 4, subfamily f, polypeptide 1 (Cyp4f15); phospholipase A2, group VII (Pla2g7); and phosphodiesterase 9a (Pde9a). This subcongenic strain offers a unique model for dissecting the contributions and possible interactions among genes controlling food intake and physical activity, key components of energy balance.


Subject(s)
Chromosomes, Mammalian , Dietary Carbohydrates/administration & dosage , Energy Intake/genetics , Motor Activity/genetics , Animals , Body Composition , Calorimetry, Indirect , Crosses, Genetic , Eating/genetics , Gene Expression Profiling , Mice , Mice, Congenic , Mice, Inbred C57BL , Motor Activity/physiology , Phenotype , Protein Array Analysis , Quantitative Trait Loci , Up-Regulation
13.
Metabolism ; 57(7 Suppl 1): S10-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18555848

ABSTRACT

Published reports of botanical action are often hampered by the lack of generalized systematic approaches or by the failure to explore mechanisms that could confirm and extend the reported observations. Choice of mouse or rat housing conditions (singly or group housed) and imposed stress during handling procedures are often variable and can contribute significantly to differences in baseline phenotypes measured across studies. Differences can also be observed in the role of the extract in either the treatment of the metabolic syndrome or roles in the regulation of the emergence of metabolic syndrome. The choice of diet used can also vary between the different studies, and diet-botanical interactions must be considered. This minireview highlights the strategies being pursued by the Botanical Research Center Animal Research Core to evaluate the in vivo phenotypes of several botanical extracts during long-term feeding studies. We describe a phenotyping strategy that promotes a more rigorous interpretation of botanical action and can suggest or eliminate possible mechanisms that may be involved. We discuss the importance of selecting the mouse model, as background strain can significantly alter the underlying susceptibilities to the various components of metabolic syndrome. Finally, we present data suggesting that one of the major botanical extracts being studied, an extract of Russian tarragon, may manifest a mouse strain genotype-specific insulin-sensitizing phenotype.


Subject(s)
Artemisia , Insulin Resistance , Metabolic Syndrome/drug therapy , Metabolic Syndrome/genetics , Plant Extracts/therapeutic use , Animals , Artemisia/chemistry , Diet , Genotype , Humans , Insulin Resistance/genetics , Metabolic Syndrome/metabolism , Mice , Models, Animal , Phytotherapy , Plant Extracts/pharmacology
14.
Eur J Pharmacol ; 585(1): 14-23, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18396272

ABSTRACT

The mu-opioid receptor encoded by the Oprm1 gene plays a crucial role in the mediation of food reward and drug-induced positive reinforcement, but its genetic deletion has been shown to provide food intake-independent, partial protection from diet-induced obesity. We hypothesized that mu-opioid receptor-deficient mice would show an even greater, intake-dependent, resistance to high-fat diet-induced obesity if the diet comprises a sweet component. We generated an F2 population by crossing the heterozygous offspring of homozygous female Oprm1(-/-) mice (on a mixed C57BL/6 and BALB/c genetic background) with male inbred C57BL/6 mice. Groups of genotyped wild-type (WT) and homozygous mutant (KO) males and females were fed either control chow or a high caloric palatable diet consisting of sweet, liquid chocolate-flavored Ensure together with a solid high-fat diet. Food intake, body weight, and body composition was measured over a period of 16 weeks. Unexpectedly, male, and to a lesser extent female, KO mice fed chow for the entire period showed progressively increased body weight and adiposity while eating significantly more chow. In contrast, when exposed to the sweet plus high-fat diet, male, and to a lesser extent female, KO mice gained significantly less body weight and fat mass compared to WT mice when using chow fed counterparts for reference values. Male KO mice consumed 33% less of the sweet liquid diet but increased intake of high-fat pellets, so that total calorie intake was not different from WT animals. These results demonstrate a dissociation of the role of mu-opioid receptors in the control of adiposity for different diets and sex. On a bland diet, normal receptor function appears to confer a slightly catabolic predisposition, but on a highly palatable diet, it confers an anabolic metabolic profile, favoring fat accretion. Because of the complexity of mu-opioid gene regulation and tissue distribution, more selective and targeted approaches will be necessary to fully understand the underlying mechanisms.


Subject(s)
Adipose Tissue/physiopathology , Dietary Fats/administration & dosage , Obesity/physiopathology , Receptors, Opioid, mu/physiology , Animals , Body Weight/physiology , Crosses, Genetic , Disease Susceptibility , Endorphins/genetics , Energy Intake , Female , Glucose/metabolism , Glucose Tolerance Test , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Obesity/genetics , Receptors, Opioid, mu/genetics , Sex Factors
15.
Metabolism ; 56(12): 1635-42, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17998014

ABSTRACT

Diets that are high in dietary fiber are reported to have substantial health benefits. We sought to compare the metabolic effects of 3 types of dietary fibers -- sugarcane fiber (SCF), psyllium (PSY), and cellulose (CEL) -- on body weight, carbohydrate metabolism, and stomach ghrelin gene expression in a high-fat diet-fed mouse model. Thirty-six male mice (C57BL/6) were randomly divided into 4 groups that consumed high-fat diet alone (HFD) or high-fat diet containing 10% SCF, PSY, and CEL, respectively. After baseline measurements were assessed for body weight, plasma insulin, glucose, leptin, and glucagon-like peptide 1 (GLP-1), animals were treated for 12 weeks. Parameters were reevaluated at the end of study. Whereas there was no difference at the baseline, body weight gains in the PSY and SCF groups were significantly lower than in the CEL group at the end of study. No difference in body weight was observed between the PSY and SCF animals. Body composition analysis demonstrated that fat mass in the SCF group was considerably lower than in the CEL and HFD groups. In addition, fasting plasma glucose and insulin and areas under the curve of intraperitoneal glucose tolerance test were also significantly lower in the SCF and PSY groups than in the CEL and HFD groups. Moreover, fasting plasma concentrations of leptin were significantly lower and GLP-1 level was 2-fold higher in the SCF and PSY mice than in the HFD and CEL mice. Ghrelin messenger RNA levels of stomach in the SCF group were significantly lower than in the CEL and HFD groups as well. These results suggest differences in response to dietary fiber intake in this animal model because high-fat diets incorporating dietary fibers such as SCF and PSY appeared to attenuate weight gain, enhance insulin sensitivity, and modulate leptin and GLP-1 secretion and gastric ghrelin gene expression.


Subject(s)
Carbohydrate Metabolism/physiology , Dietary Fiber/administration & dosage , Ghrelin/biosynthesis , Stomach/physiology , Weight Gain/physiology , Animals , Blood Glucose/metabolism , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Dietary Fiber/metabolism , Eating/physiology , Gastric Mucosa/metabolism , Ghrelin/genetics , Glucagon-Like Peptide 1/blood , Insulin/blood , Leptin/blood , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
16.
Exp Dermatol ; 12(3): 245-54, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12823437

ABSTRACT

Harlequin ichthyosis (HI) is a rare and usually fatal scaling skin disorder. The HI mutant mouse (ichq/ichq) has many similarities to the human disorder and provides an important model to identify candidate genes. In this study, we report refined mapping of the mouse ichq locus and consideration of the candidate genes: calpain 1 (Capn1), phospholipase C beta 3 (Plcb3), and Rela and Ikka/Chuk that encode components of the nuclear factor-kappa B (NF-kappaB) pathway. Each are strong candidates because of epidermal expression and/or changes in expression in human HI. All candidates are linked to the ichq locus on mouse Chromosome 19, although Ikka is located more distally. Genetic mapping in mouse has narrowed the ichq critical region to 4 cM. Keratinocytes from skin of +/+, +/ichq and ichq/ichq mice were cultured; all genotypes had similar expression of epidermal differentiation markers. RT-PCR amplification and sequence analysis of each candidate gene did not reveal any mutations in the ichq mouse. Mutational screening of CAPN1 cDNA from different human HI cases revealed a R433P change, but analysis of 50 normal samples demonstrated that this was an apparent polymorphism. Sequence of RELA in five unrelated human HI cases was normal. The results provide compelling evidence that none of these genes are the primary defect in the ichq mouse and that CAPN1 and RELA are not mutated in the human disorder.


Subject(s)
Epidermis/physiopathology , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/physiopathology , Keratinocytes/physiology , Amino Acid Sequence , Animals , Calpain/genetics , Cell Differentiation , Cells, Cultured , Chromosome Mapping , Cytoplasm/physiology , Disease Models, Animal , Epidermis/pathology , Gene Expression , Genetic Markers , Humans , I-kappa B Kinase , Keratinocytes/cytology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Molecular Sequence Data , NF-kappa B/genetics , Pedigree , Protein Serine-Threonine Kinases/genetics , Transcription Factor RelA
17.
Nat Genet ; 30(4): 401-5, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11925566

ABSTRACT

Once a mutation in the gene tub was identified as the cause of obesity, retinal degeneration and hearing loss in tubby mice, it became increasingly evident that the members of the tub gene family (tulps) influence maintenance and function of the neuronal cell lineage. Suggested molecular functions of tubby-like proteins include roles in vesicular trafficking, mediation of insulin signaling and gene transcription. The mechanisms through which tub functions in neurons, however, have yet to be elucidated. Here we report the positional cloning of an auditory quantitative trait locus (QTL), the modifier of tubby hearing 1 gene (moth1), whose wildtype alleles from strains AKR/J, CAST/Ei and 129P2/OlaHsd protect tubby mice from hearing loss. Through a transgenic rescue experiment, we verified that sequence polymorphisms in the neuron-specific microtubule-associated protein 1a gene (Mtap1a) observed in the susceptible strain C57BL/6J (B6) are crucial for the hearing-loss phenotype. We also show that these polymorphisms change the binding efficiency of MTAP1A to postsynaptic density molecule 95 (PSD95), a core component in the cytoarchitecture of synapses. This indicates that at least some of the observed polymorphisms are functionally important and that the hearing loss in C57BL/6J-tub/tub (B6-tub/tub) mice may be caused by impaired protein interactions involving MTAP1A. We therefore propose that tub may be associated with synaptic function in neuronal cells.


Subject(s)
Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Proteins/genetics , Adaptor Proteins, Signal Transducing , Alleles , Animals , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Disks Large Homolog 4 Protein , Gene Library , Genetic Markers , Guanylate Kinases , Immunoblotting , Insulin/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Models, Genetic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phenotype , Polymorphism, Genetic , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Quantitative Trait, Heritable , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Synapses/metabolism , Transcription, Genetic , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...