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










Publication year range
1.
J Mol Cell Cardiol ; 156: 45-56, 2021 07.
Article in English | MEDLINE | ID: mdl-33773996

ABSTRACT

CRELD1 (Cysteine-Rich with EGF-Like Domains 1) is a risk gene for non-syndromic atrioventricular septal defects in human patients. In a mouse model, Creld1 has been shown to be essential for heart development, particularly in septum and valve formation. However, due to the embryonic lethality of global Creld1 knockout (KO) mice, its cell type-specific function during peri- and postnatal stages remains unknown. Here, we generated conditional Creld1 KO mice lacking Creld1 either in the endocardium (KOTie2) or the myocardium (KOMyHC). Using a combination of cardiac phenotyping, histology, immunohistochemistry, RNA-sequencing, and flow cytometry, we demonstrate that Creld1 function in the endocardium is dispensable for heart development. Lack of myocardial Creld1 causes extracellular matrix remodeling and trabeculation defects by modulation of the Notch1 signaling pathway. Hence, KOMyHC mice die early postnatally due to myocardial hypoplasia. Our results reveal that Creld1 not only controls the formation of septa and valves at an early stage during heart development, but also cardiac maturation and function at a later stage. These findings underline the central role of Creld1 in mammalian heart development and function.


Subject(s)
Cell Adhesion Molecules/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiology , Myocardium/metabolism , Organogenesis/genetics , Animals , Biomarkers , Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Mice, Knockout , Single-Cell Analysis
2.
Mamm Genome ; 31(1-2): 30-48, 2020 02.
Article in English | MEDLINE | ID: mdl-32060626

ABSTRACT

The collaborative cross (CC) is a large panel of mouse-inbred lines derived from eight founder strains (NOD/ShiLtJ, NZO/HILtJ, A/J, C57BL/6J, 129S1/SvImJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ). Here, we performed a comprehensive and comparative phenotyping screening to identify phenotypic differences and similarities between the eight founder strains. In total, more than 300 parameters including allergy, behavior, cardiovascular, clinical blood chemistry, dysmorphology, bone and cartilage, energy metabolism, eye and vision, immunology, lung function, neurology, nociception, and pathology were analyzed; in most traits from sixteen females and sixteen males. We identified over 270 parameters that were significantly different between strains. This study highlights the value of the founder and CC strains for phenotype-genotype associations of many genetic traits that are highly relevant to human diseases. All data described here are publicly available from the mouse phenome database for analyses and downloads.


Subject(s)
Mice, Inbred Strains/genetics , Phenotype , Animals , Collaborative Cross Mice/genetics , Databases, Genetic , Female , Genetic Association Studies , Genotype , Male , Mice , Quantitative Trait Loci , Species Specificity
3.
Matrix Biol ; 77: 4-22, 2019 04.
Article in English | MEDLINE | ID: mdl-29777767

ABSTRACT

Sterile inflammation is a therapeutic target in many diseases where it represents an important initiator of disease progression. However, the detailed mechanisms underlying its evolution and biological relevance are not yet completely elucidated. Biglycan, a prototype extracellular matrix-derived damage-associated molecular pattern, mediates sterile inflammation in macrophages through Toll-like receptor (TLR) 2 and/or TLR4-dependent signaling pathways. Here we discovered that soluble biglycan is a novel high-affinity ligand for CD14, a well-known GPI-anchored co-receptor for TLRs. CD14 is required for all biglycan-mediated TLR2/4 dependent inflammatory signaling pathways in macrophages. By binding to CD14 and choosing different TLR signaling branches, biglycan induced TNF-α and CCL2 via TLR2/4, HSP70 through TLR2, and CCL5 via TLR4. Mechanistically, biglycan evoked phosphorylation and subsequent nuclear translocation of p38, p44/42, and NF-κB, and these effects were due to a specific, high-affinity interaction between biglycan protein core and CD14. Finally, we provide proof-of-principle for the requirement of CD14, by transiently overexpressing biglycan in a mouse model of renal ischemia/reperfusion injury performed in Cd14-/- mice. Lack of Cd14 prevented biglycan-mediated cytokine expression, recruitment of macrophages, M1 macrophage polarization as well as mitigated the tubular damage and serum creatinine levels, thereby improving renal function. Thus, CD14 inhibition could lead to the reduction in the activation of biglycan-TLR2/4 signaling pathways and could be a novel therapeutic approach in inflammatory kidney diseases.


Subject(s)
Biglycan/pharmacology , Kidney/drug effects , Lipopolysaccharide Receptors/genetics , Macrophages/drug effects , Reperfusion Injury/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Biglycan/immunology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Extracellular Matrix/chemistry , Extracellular Matrix/immunology , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , Humans , Inflammation , Kidney/immunology , Kidney/pathology , Ligands , Lipopolysaccharide Receptors/deficiency , Lipopolysaccharide Receptors/immunology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Protein Binding , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Signal Transduction , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
4.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2083-2093, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30557699

ABSTRACT

Mutations in the X chromosomal tRNA 2'­O­methyltransferase FTSJ1 cause intellectual disability (ID). Although the gene is ubiquitously expressed affected individuals present no consistent clinical features beyond ID. In order to study the pathological mechanism involved in the aetiology of FTSJ1 deficiency-related cognitive impairment, we generated and characterized an Ftsj1 deficient mouse line based on the gene trapped stem cell line RRD143. Apart from an impaired learning capacity these mice presented with several statistically significantly altered features related to behaviour, pain sensing, bone and energy metabolism, the immune and the hormone system as well as gene expression. These findings show that Ftsj1 deficiency in mammals is not phenotypically restricted to the brain but affects various organ systems. Re-examination of ID patients with FTSJ1 mutations from two previously reported families showed that several features observed in the mouse model were recapitulated in some of the patients. Though the clinical spectrum related to Ftsj1 deficiency in mouse and man is variable, we suggest that an increased pain threshold may be more common in patients with FTSJ1 deficiency. Our findings demonstrate novel roles for Ftsj1 in maintaining proper cellular and tissue functions in a mammalian organism.


Subject(s)
Disease Models, Animal , Intellectual Disability/etiology , Mental Retardation, X-Linked/genetics , Methyltransferases/physiology , Mutation , Nuclear Proteins/genetics , tRNA Methyltransferases/physiology , Animals , Behavior, Animal , Cognition Disorders/etiology , Cognition Disorders/pathology , Family , Female , Intellectual Disability/pathology , Male , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptive Pain/etiology , Nociceptive Pain/pathology , Nuclear Proteins/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
5.
EMBO J ; 37(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30087110

ABSTRACT

Epitranscriptomic events such as adenosine-to-inosine (A-to-I) RNA editing by ADAR can recode mRNAs to translate novel proteins. Editing of the mRNA that encodes actin crosslinking protein Filamin A (FLNA) mediates a Q-to-R transition in the interactive C-terminal region. While FLNA editing is conserved among vertebrates, its physiological function remains unclear. Here, we show that cardiovascular tissues in humans and mice show massive editing and that FLNA RNA is the most prominent substrate. Patient-derived RNA-Seq data demonstrate a significant drop in FLNA editing associated with cardiovascular diseases. Using mice with only impaired FLNA editing, we observed increased vascular contraction and diastolic hypertension accompanied by increased myosin light chain phosphorylation, arterial remodeling, and left ventricular wall thickening, which eventually causes cardiac remodeling and reduced systolic output. These results demonstrate a causal relationship between RNA editing and the development of cardiovascular disease indicating that a single epitranscriptomic RNA modification can maintain cardiovascular health.


Subject(s)
Blood Pressure , Filamins/metabolism , Hypertension/metabolism , Muscle Contraction , Myocardium/metabolism , RNA Editing , RNA Precursors/metabolism , Vascular Remodeling , Animals , Filamins/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Hypertension/genetics , Hypertension/pathology , Mice , Myocardium/pathology , RNA Precursors/genetics , Sequence Analysis, RNA
6.
Biochem Biophys Res Commun ; 503(4): 2770-2777, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30100055

ABSTRACT

Heterozygous missense mutations in the human VCP gene cause inclusion body myopathy associated with Paget disease of bone and fronto-temporal dementia (IBMPFD) and amyotrophic lateral sclerosis (ALS). The exact molecular mechanisms by which VCP mutations cause disease manifestation in different tissues are incompletely understood. In the present study, we report the comprehensive analysis of a newly generated R155C VCP knock-in mouse model, which expresses the ortholog of the second most frequently occurring human pathogenic VCP mutation. Heterozygous R155C VCP knock-in mice showed decreased plasma lactate, serum albumin and total protein concentrations, platelet numbers, and liver to body weight ratios, and increased oxygen consumption and CD8+/Ly6C + T-cell fractions, but none of the typical human IBMPFD or ALS pathologies. Breeding of heterozygous mice did not yield in the generation of homozygous R155C VCP knock-in animals. Immunoblotting showed identical total VCP protein levels in human IBMPFD and murine R155C VCP knock-in tissues as compared to wild-type controls. However, while in human IBMPFD skeletal muscle tissue 70% of the total VCP mRNA was derived from the mutant allele, in R155C VCP knock-in mice only 5% and 7% mutant mRNA were detected in skeletal muscle and brain tissue, respectively. The lack of any obvious IBMPFD or ALS pathology could thus be a consequence of the very low expression of mutant VCP. We conclude that the increased and decreased fractions of the R155C mutant VCP mRNA in man and mice, respectively, are due to missense mutation-induced, divergent alterations in the biological half-life of the human and murine mutant mRNAs. Furthermore, our work suggests that therapy approaches lowering the expression of the mutant VCP mRNA below a critical threshold may ameliorate the intrinsic disease pathology.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Genes, Lethal , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Myositis, Inclusion Body/genetics , Osteitis Deformans/genetics , Valosin Containing Protein/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Brain/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Gene Expression Regulation , Gene Knock-In Techniques , Heterozygote , Humans , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Osteitis Deformans/metabolism , Osteitis Deformans/pathology , Signal Transduction , Species Specificity , Valosin Containing Protein/metabolism
7.
JCI Insight ; 3(7)2018 04 05.
Article in English | MEDLINE | ID: mdl-29618659

ABSTRACT

Mesenchymal TNF signaling is etiopathogenic for inflammatory diseases such as rheumatoid arthritis and spondyloarthritis (SpA). The role of Tnfr1 in arthritis has been documented; however, Tnfr2 functions are unknown. Here, we investigate the mesenchymal-specific role of Tnfr2 in the TnfΔARE mouse model of SpA in arthritis and heart valve stenosis comorbidity by cell-specific, Col6a1-cre-driven gene targeting. We find that TNF/Tnfr2 signaling in resident synovial fibroblasts (SFs) and valvular interstitial cells (VICs) is detrimental for both pathologies, pointing to common cellular mechanisms. In contrast, systemic Tnfr2 provides protective signaling, since its complete deletion leads to severe deterioration of both pathologies. SFs and VICs lacking Tnfr2 fail to acquire pathogenic activated phenotypes and display increased expression of antiinflammatory cytokines associated with decreased Akt signaling. Comparative RNA sequencing experiments showed that the majority of the deregulated pathways in TnfΔARE mesenchymal-origin SFs and VICs, including proliferation, inflammation, migration, and disease-specific genes, are regulated by Tnfr2; thus, in its absence, they are maintained in a quiescent nonpathogenic state. Our data indicate a pleiotropy of Tnfr2 functions, with mesenchymal Tnfr2 driving cell activation and arthritis/valve stenosis pathogenesis only in the presence of systemic Tnfr2, whereas nonmesenchymal Tnfr2 overcomes this function, providing protective signals and, thus, containing both pathologies.


Subject(s)
Aortic Valve Stenosis/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Signal Transduction/immunology , Spondylarthritis/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Spondylarthritis/complications , Spondylarthritis/genetics , Spondylarthritis/pathology , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
PLoS Biol ; 16(4): e2005019, 2018 04.
Article in English | MEDLINE | ID: mdl-29659570

ABSTRACT

Animal welfare requires the adequate housing of animals to ensure health and well-being. The application of environmental enrichment is a way to improve the well-being of laboratory animals. However, it is important to know whether these enrichment items can be incorporated in experimental mouse husbandry without creating a divide between past and future experimental results. Previous small-scale studies have been inconsistent throughout the literature, and it is not yet completely understood whether and how enrichment might endanger comparability of results of scientific experiments. Here, we measured the effect on means and variability of 164 physiological parameters in 3 conditions: with nesting material with or without a shelter, comparing these 2 conditions to a "barren" regime without any enrichments. We studied a total of 360 mice from each of 2 mouse strains (C57BL/6NTac and DBA/2NCrl) and both sexes for each of the 3 conditions. Our study indicates that enrichment affects the mean values of some of the 164 parameters with no consistent effects on variability. However, the influence of enrichment appears negligible compared to the effects of other influencing factors. Therefore, nesting material and shelters may be used to improve animal welfare without impairment of experimental outcome or loss of comparability to previous data collected under barren housing conditions.


Subject(s)
Animal Welfare/ethics , Environment, Controlled , Nesting Behavior/physiology , Animal Welfare/economics , Animals , Energy Metabolism/physiology , Female , Heart Function Tests/methods , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nociception/physiology
9.
Behav Brain Res ; 352: 187-196, 2018 10 15.
Article in English | MEDLINE | ID: mdl-28966146

ABSTRACT

Since decades, model organisms have provided an important approach for understanding the mechanistic basis of human diseases. The German Mouse Clinic (GMC) was the first phenotyping facility that established a collaboration-based platform for phenotype characterization of mouse lines. In order to address individual projects by a tailor-made phenotyping strategy, the GMC advanced in developing a series of pipelines with tests for the analysis of specific disease areas. For a general broad analysis, there is a screening pipeline that covers the key parameters for the most relevant disease areas. For hypothesis-driven phenotypic analyses, there are thirteen additional pipelines with focus on neurological and behavioral disorders, metabolic dysfunction, respiratory system malfunctions, immune-system disorders and imaging techniques. In this article, we give an overview of the pipelines and describe the scientific rationale behind the different test combinations.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Phenotype , Animals , Humans
10.
J Biomed Sci ; 24(1): 57, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28818080

ABSTRACT

BACKGROUND: Increased levels of blood plasma urea were used as phenotypic parameter for establishing novel mouse models for kidney diseases on the genetic background of C3H inbred mice in the phenotype-driven Munich ENU mouse mutagenesis project. The phenotypically dominant mutant line HST014 was established and further analyzed. METHODS: Analysis of the causative mutation as well as the standardized, systemic phenotypic analysis of the mutant line was carried out. RESULTS: The causative mutation was detected in the potassium channel tetramerization domain containing 1 (Kctd1) gene which leads to the amino acid exchange Kctd1 I27N thereby affecting the functional BTB domain of the protein. This line is the first mouse model harboring a Kctd1 mutation. Kctd1 I27N homozygous mutant mice die perinatally. Standardized, systemic phenotypic analysis of Kctd1 I27N heterozygous mutants was carried out in the German Mouse Clinic (GMC). Systematic morphological investigation of the external physical appearance did not detect the specific alterations that are described in KCTD1 mutant human patients affected by the scalp-ear-nipple (SEN) syndrome. The main pathological phenotype of the Kctd1 I27N heterozygous mutant mice consists of kidney dysfunction and secondary effects thereof, without gross additional primary alterations in the other phenotypic parameters analyzed. Genome-wide transcriptome profiling analysis at the age of 4 months revealed about 100 differentially expressed genes (DEGs) in kidneys of Kctd1 I27N heterozygous mutants as compared to wild-type controls. CONCLUSIONS: In summary, the main alteration of the Kctd1 I27N heterozygous mutants consists in kidney dysfunction. Additional analyses in 9-21 week-old heterozygous mutants revealed only few minor effects.


Subject(s)
Co-Repressor Proteins/genetics , Disease Models, Animal , Kidney Diseases/genetics , Kidney/physiopathology , Mice , Mutation , Animals , Female , Male , Mice, Inbred C3H , Phenotype
11.
Nat Commun ; 8(1): 155, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28761067

ABSTRACT

Dietary restriction regimes extend lifespan in various animal models. Here we show that longevity in male C57BL/6J mice subjected to every-other-day feeding is associated with a delayed onset of neoplastic disease that naturally limits lifespan in these animals. We compare more than 200 phenotypes in over 20 tissues in aged animals fed with a lifelong every-other-day feeding or ad libitum access to food diet to determine whether molecular, cellular, physiological and histopathological aging features develop more slowly in every-other-day feeding mice than in controls. We also analyze the effects of every-other-day feeding on young mice on shorter-term every-other-day feeding or ad libitum to account for possible aging-independent restriction effects. Our large-scale analysis reveals overall only limited evidence for a retardation of the aging rate in every-other-day feeding mice. The data indicate that every-other-day feeding-induced longevity is sufficiently explained by delays in life-limiting neoplastic disorders and is not associated with a more general slowing of the aging process in mice.Dietary restriction can extend the life of various model organisms. Here, Xie et al. show that intermittent periods of fasting achieved through every-other-day feeding protect mice against neoplastic disease but do not broadly delay organismal aging in animals.


Subject(s)
Aging , Food Deprivation , Longevity , Animals , Male , Mice , Mice, Inbred C57BL
12.
Mol Neurobiol ; 54(10): 8242-8262, 2017 12.
Article in English | MEDLINE | ID: mdl-27914009

ABSTRACT

Stress experience modulates behavior, metabolism, and energy expenditure of organisms. One molecular hallmark of an acute stress response is a rapid induction of immediate early genes (IEGs) such as c-Fos and Egr family members. IEG transcription in neurons is mediated by the neuronal activity-driven gene regulator serum response factor (SRF). We show a first role of SRF in immediate and long-lasting acute restraint stress (AS) responses. For this, we employed a standardized mouse phenotyping protocol at the German Mouse Clinic (GMC) including behavioral, metabolic, and cardiologic tests as well as gene expression profiling to analyze the consequences of forebrain-specific SRF deletion in mice exposed to AS. Adult mice with an SRF deletion in glutamatergic neurons (Srf; CaMKIIa-CreERT2 ) showed hyperactivity, decreased anxiety, and impaired working memory. In response to restraint AS, instant stress reactivity including locomotor behavior and corticosterone induction was impaired in Srf mutant mice. Interestingly, even several weeks after previous AS exposure, SRF-deficient mice showed long-lasting AS-associated changes including altered locomotion, metabolism, energy expenditure, and cardiovascular changes. This suggests a requirement of SRF for mediating long-term stress coping mechanisms in wild-type mice. SRF ablation decreased AS-mediated IEG induction and activity of the actin severing protein cofilin. In summary, our data suggest an SRF function in immediate AS reactions and long-term post-stress-associated coping mechanisms.


Subject(s)
Adaptation, Psychological/physiology , Genes, Immediate-Early/physiology , Prosencephalon/metabolism , Serum Response Factor/deficiency , Stress, Psychological/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prosencephalon/pathology , Serum Response Factor/genetics , Stress, Psychological/genetics , Stress, Psychological/pathology , Time Factors
13.
Glycoconj J ; 34(3): 393-404, 2017 06.
Article in English | MEDLINE | ID: mdl-27600268

ABSTRACT

Secondary polycythemia, a disease characterized by a selective increase in circulating mature erythrocytes, is caused by enhanced erythropoietin (Epo) concentrations triggered by hypoxia-inducible factor-2α (HIF-2α). While mechanisms of hypoxia-dependent stabilization of HIF-2α protein are well established, data regarding oxygen-independent regulation of HIF-2α are sparse. In this study, we generated a novel transgenic mouse model, in which biglycan was constitutively overexpressed and secreted by hepatocytes (BGN Tg), thereby providing a constant source of biglycan released into the blood stream. We discovered that although the mice were apparently normal, they harbored an increase in mature circulating erythrocytes. In addition to erythrocytosis, the BGN Tg mice showed elevated hemoglobin concentrations, hematocrit values and enhanced total iron binding capacity, revealing a clinical picture of polycythemia. In BGN Tg mice markedly enhanced Epo mRNA expression was observed in the liver and kidney, while elevated Epo protein levels were found in liver, kidney and blood. Mechanistically, we showed that the transgenic animals had an abundance of HIF-2α protein in the liver and kidney. Finally, by transiently overexpressing circulating biglycan in mice deficient in various Toll-like receptors (TLRs), we determined that this novel function of biglycan to promote Epo synthesis was specifically mediated by a selective interaction with TLR2. Thus, we discovered a novel biological pathway of soluble biglycan inducing HIF-2α protein stabilization and Epo production presumably in an oxygen-independent manner, ultimately giving rise to secondary polycythemia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Biglycan/genetics , Erythropoietin/genetics , Hepatocytes/metabolism , Polycythemia/genetics , RNA, Messenger/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biglycan/metabolism , Disease Models, Animal , Erythrocyte Count , Erythrocytes/metabolism , Erythrocytes/pathology , Erythropoietin/biosynthesis , Gene Expression Regulation , Hematocrit , Hemoglobins/metabolism , Hepatocytes/pathology , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Polycythemia/metabolism , Polycythemia/pathology , RNA, Messenger/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
14.
G3 (Bethesda) ; 6(12): 4035-4046, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27815347

ABSTRACT

The vertebrate Scube (Signal peptide, CUB, and EGF-like domain-containing protein) family consists of three independent members, Scube1-3, which encode secreted cell surface-associated membrane glycoproteins. Limited information about the general function of this gene family is available, and their roles during adulthood. Here, we present the first Scube3 mutant mouse line (Scube3N294K/N294K), which clearly shows phenotypic alterations by carrying a missense mutation in exon 8, and thus contributes to our understanding of SCUBE3 functions. We performed a detailed phenotypic characterization in the German Mouse Clinic (GMC). Scube3N294K/N294K mutants showed morphological abnormalities of the skeleton, alterations of parameters relevant for bone metabolism, changes in renal function, and hearing impairments. These findings correlate with characteristics of the rare metabolic bone disorder Paget disease of bone (PDB), associated with the chromosomal region of human SCUBE3 In addition, alterations in energy metabolism, behavior, and neurological functions were detected in Scube3N294K/N294K mice. The Scube3N294K/N294K mutant mouse line may serve as a new model for further studying the effect of impaired SCUBE3 gene function.


Subject(s)
Genetic Association Studies , Glycoproteins/genetics , Mutation , Phenotype , Animals , Bone and Bones/metabolism , Calcium-Binding Proteins , Chromosome Mapping , Disease Models, Animal , Energy Metabolism/genetics , Exome , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Kidney/metabolism , Kidney/physiopathology , Kidney Function Tests , Male , Mice , Mice, Knockout , Osteitis Deformans/genetics , Osteitis Deformans/metabolism , Osteitis Deformans/pathology , Skeleton/abnormalities
15.
Nat Med ; 22(12): 1428-1438, 2016 12.
Article in English | MEDLINE | ID: mdl-27841876

ABSTRACT

Aging is associated with an increased risk of cardiovascular disease and death. Here we show that oral supplementation of the natural polyamine spermidine extends the lifespan of mice and exerts cardioprotective effects, reducing cardiac hypertrophy and preserving diastolic function in old mice. Spermidine feeding enhanced cardiac autophagy, mitophagy and mitochondrial respiration, and it also improved the mechano-elastical properties of cardiomyocytes in vivo, coinciding with increased titin phosphorylation and suppressed subclinical inflammation. Spermidine feeding failed to provide cardioprotection in mice that lack the autophagy-related protein Atg5 in cardiomyocytes. In Dahl salt-sensitive rats that were fed a high-salt diet, a model for hypertension-induced congestive heart failure, spermidine feeding reduced systemic blood pressure, increased titin phosphorylation and prevented cardiac hypertrophy and a decline in diastolic function, thus delaying the progression to heart failure. In humans, high levels of dietary spermidine, as assessed from food questionnaires, correlated with reduced blood pressure and a lower incidence of cardiovascular disease. Our results suggest a new and feasible strategy for protection against cardiovascular disease.


Subject(s)
Aging/drug effects , Autophagy/drug effects , Blood Pressure/drug effects , Heart/drug effects , Longevity/drug effects , Mitochondria, Heart/drug effects , Mitophagy/drug effects , Myocytes, Cardiac/drug effects , Spermidine/pharmacology , Adult , Aged , Aging/immunology , Aging/metabolism , Animals , Autophagy-Related Protein 5/genetics , Cardiomegaly/diagnostic imaging , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/epidemiology , Chromatography, High Pressure Liquid , Connectin/drug effects , Connectin/metabolism , Cytokines/drug effects , Cytokines/immunology , Diastole , Diet/statistics & numerical data , Echocardiography , Female , Gene Expression/drug effects , Glucose Tolerance Test , Heart/diagnostic imaging , Heart Failure , Humans , Immunoblotting , Inflammation , Male , Mass Spectrometry , Mice , Middle Aged , Mitochondria, Heart/metabolism , Phosphorylation/drug effects , Prospective Studies , Rats , Rats, Inbred Dahl , Surveys and Questionnaires
16.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27720451

ABSTRACT

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Subject(s)
Glucagon/therapeutic use , Metabolic Diseases/drug therapy , Triiodothyronine/drug effects , Animals , Atherosclerosis/drug therapy , Body Weight/drug effects , Bone and Bones/drug effects , Chemical Engineering/methods , Cholesterol/metabolism , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Drug Combinations , Drug Delivery Systems , Drug Synergism , Glucagon/adverse effects , Glucagon/chemistry , Glucagon/pharmacology , Hyperglycemia/drug therapy , Liver/drug effects , Liver/metabolism , Mice , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Triiodothyronine/adverse effects , Triiodothyronine/chemistry , Triiodothyronine/pharmacology
17.
Basic Res Cardiol ; 111(1): 6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26659360

ABSTRACT

Cardiomyopathy is one of the most common causes of chronic heart failure worldwide. Mutations in the gene encoding nexilin (NEXN) occur in patients with both hypertrophic and dilated cardiomyopathy (DCM); however, little is known about the pathophysiological mechanisms and relevance of NEXN to these disorders. Here, we evaluated the functional role of NEXN using a constitutive Nexn knock-out (KO) mouse model. Heterozygous (Het) mice were inter-crossed to produce wild-type (WT), Het, and homozygous KO mice. At birth, 32, 46, and 22 % of the mice were WT, Het, and KO, respectively, which is close to the expected Mendelian ratio. After postnatal day 6, the survival of the Nexn KO mice decreased dramatically and all of the animals died by day 8. Phenotypic characterizations of the WT and KO mice were performed at postnatal days 1, 2, 4, and 6. At birth, the relative heart weights of the WT and KO mice were similar; however, at day 4, the relative heart weight of the KO group was 2.3-fold higher than of the WT group. In addition, the KO mice developed rapidly progressive cardiomyopathy with left ventricular dilation and wall thinning and decreased cardiac function. At day 6, the KO mice developed a fulminant DCM phenotype characterized by dilated ventricular chambers and systolic dysfunction. At this stage, collagen deposits and some elastin deposits were observed within the left ventricle cavity, which resembles the features of endomyocardial fibroelastosis (EFE). Overall, these results further emphasize the role of NEXN in DCM and suggest a novel role in EFE.


Subject(s)
Cardiomyopathies/metabolism , Endocardial Fibroelastosis/metabolism , Microfilament Proteins/deficiency , Animals , Blotting, Western , Cardiomyopathies/pathology , Disease Models, Animal , Echocardiography , Endocardial Fibroelastosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Polymerase Chain Reaction
19.
Int J Cardiovasc Imaging ; 31(4): 669-79, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25627778

ABSTRACT

Consistent protocols for the assessment of diastolic and systolic cardiac function to assure the comparability of existing data on preclinical models are missing. Calcineurin transgene (CN) mice are a preclinical model for hypertrophic and failing hearts. We aimed at evaluating left and right ventricular structural and functional remodeling in CN hearts with an optimized phenotyping protocol. We developed a protocol using techniques and indices comparable to those from human diagnostics for comprehensive in vivo cardiac screening using high-frequency echocardiography, Doppler, electrocardiography and cardiac magnetic resonance (CMR) techniques. We measured left and right ventricular dimensions and function, pulmonary and mitral flow pattern and the hearts electrophysiology non-invasively in <1 h per mouse. We found severe biventricular dilation and a drastic decline in performance in accordance with a condition of heart failure (HF), diastolic dysfunction and defects in electrical conduction in 8-week-old calcineurin transgenic mice. Echocardiography of the left ventricle was performed with and without anesthesia. In all cases absolute values on echocardiography compared with CMR were smaller for LV dimension and wall thickness, resulting in higher fractional shorting and ejection fraction. The study protocol described here opens opportunities to assess the added value of combined echocardiography, Doppler, CMR and ECG recording techniques for the diagnosis of biventricular cardiac pathologies i.e. of HF and to study symptom occurrence and disease progression non-invasively in high-throughput. Phenotyping CN hearts revealed new symptom occurrence and allowed insights into the diverse phenotype of hypertrophic failing hearts.


Subject(s)
Calcineurin/genetics , Cardiomyopathy, Hypertrophic/diagnosis , Heart Failure/diagnosis , High-Throughput Screening Assays/methods , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Right Ventricular/diagnosis , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Right/diagnosis , Animals , Calcineurin/metabolism , Cardiomyopathy, Hypertrophic/enzymology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Disease Models, Animal , Disease Progression , Echocardiography, Doppler , Electrocardiography , Female , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/physiopathology , Hemodynamics , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Mice, Transgenic , Myosin Heavy Chains/genetics , Phenotype , Predictive Value of Tests , Promoter Regions, Genetic , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Right/enzymology , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Left , Ventricular Function, Right , Ventricular Myosins/genetics , Ventricular Remodeling
20.
PLoS One ; 9(8): e104568, 2014.
Article in English | MEDLINE | ID: mdl-25111779

ABSTRACT

The transcription factor Zscan10 had been attributed a role as a pluripotency factor in embryonic stem cells based on its interaction with Oct4 and Sox2 in in vitro assays. Here we suggest a potential role of Zscan10 in controlling progenitor cell populations in vivo. Mice homozygous for a Zscan10 mutation exhibit reduced weight, mild hypoplasia in the spleen, heart and long bones and phenocopy an eye malformation previously described for Sox2 hypomorphs. Phenotypic abnormalities are supported by the nature of Zscan10 expression in midgestation embryos and adults suggesting a role for Zscan10 in either maintaining progenitor cell subpopulation or impacting on fate choice decisions thereof.


Subject(s)
Genetic Pleiotropy , Transcription Factors/genetics , Animals , Behavior, Animal , Body Weight/genetics , Bone Density/genetics , Bone and Bones/physiology , Codon, Initiator/genetics , Eye/growth & development , Female , Gene Expression Regulation/genetics , Homozygote , Male , Mice , Mutation , Organ Size/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pregnancy , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL
...