Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 4481, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37491352

ABSTRACT

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.


Subject(s)
COVID-19 , Interferon Type I , Humans , Mice , Animals , Cytokines , SARS-CoV-2 , Mice, Transgenic , Inflammation/genetics , Disease Models, Animal , Lung
3.
Nature ; 537(7621): 508-514, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27626380

ABSTRACT

Approximately one-third of all mammalian genes are essential for life. Phenotypes resulting from knockouts of these genes in mice have provided tremendous insight into gene function and congenital disorders. As part of the International Mouse Phenotyping Consortium effort to generate and phenotypically characterize 5,000 knockout mouse lines, here we identify 410 lethal genes during the production of the first 1,751 unique gene knockouts. Using a standardized phenotyping platform that incorporates high-resolution 3D imaging, we identify phenotypes at multiple time points for previously uncharacterized genes and additional phenotypes for genes with previously reported mutant phenotypes. Unexpectedly, our analysis reveals that incomplete penetrance and variable expressivity are common even on a defined genetic background. In addition, we show that human disease genes are enriched for essential genes, thus providing a dataset that facilitates the prioritization and validation of mutations identified in clinical sequencing efforts.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Genes, Essential/genetics , Genes, Lethal/genetics , Mutation/genetics , Phenotype , Animals , Conserved Sequence/genetics , Disease , Genome-Wide Association Study , High-Throughput Screening Assays , Humans , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Mice, Knockout , Penetrance , Polymorphism, Single Nucleotide/genetics , Sequence Homology
4.
Nat Genet ; 47(11): 1260-3, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437028

ABSTRACT

Heterotaxy results from a failure to establish normal left-right asymmetry early in embryonic development. By whole-exome sequencing, whole-genome sequencing and high-throughput cohort resequencing, we identified recessive mutations in MMP21 (encoding matrix metallopeptidase 21) in nine index cases with heterotaxy. In addition, Mmp21-mutant mice and mmp21-morphant zebrafish displayed heterotaxy and abnormal cardiac looping, respectively, suggesting a new role for extracellular matrix remodeling in the establishment of laterality in vertebrates.


Subject(s)
Body Patterning/genetics , Heterotaxy Syndrome/genetics , Matrix Metalloproteinases, Secreted/genetics , Point Mutation , Vertebrates/genetics , Animals , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Family Health , Female , Gene Expression Regulation, Developmental , Genes, Recessive , Heart/embryology , Heart Defects, Congenital/genetics , Humans , In Situ Hybridization , Male , Mice , Pedigree , Sequence Analysis, DNA/methods , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Am J Physiol Endocrinol Metab ; 308(5): E402-13, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25516547

ABSTRACT

As development proceeds from the embryonic to fetal stages, cardiac energy demands increase substantially, and oxidative phosphorylation of ADP to ATP in mitochondria becomes vital. Relatively little, however, is known about the signaling mechanisms regulating the transition from anaerobic to aerobic metabolism that occurs during the embryonic period. The main objective of this study was to test the hypothesis that adrenergic hormones provide critical stimulation of energy metabolism during embryonic/fetal development. We examined ATP and ADP concentrations in mouse embryos lacking adrenergic hormones due to targeted disruption of the essential dopamine ß-hydroxylase (Dbh) gene. Embryonic ATP concentrations decreased dramatically, whereas ADP concentrations rose such that the ATP/ADP ratio in the adrenergic-deficient group was nearly 50-fold less than that found in littermate controls by embryonic day 11.5. We also found that cardiac extracellular acidification and oxygen consumption rates were significantly decreased, and mitochondria were significantly larger and more branched in adrenergic-deficient hearts. Notably, however, the mitochondria were intact with well-formed cristae, and there was no significant difference observed in mitochondrial membrane potential. Maternal administration of the adrenergic receptor agonists isoproterenol or l-phenylephrine significantly ameliorated the decreases in ATP observed in Dbh-/- embryos, suggesting that α- and ß-adrenergic receptors were effective modulators of ATP concentrations in mouse embryos in vivo. These data demonstrate that adrenergic hormones stimulate cardiac energy metabolism during a critical period of embryonic development.


Subject(s)
Adrenergic Agents/pharmacology , Autonomic Nervous System Diseases/embryology , Autonomic Nervous System Diseases/physiopathology , Dopamine beta-Hydroxylase/deficiency , Dopamine beta-Hydroxylase/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Heart Diseases , Norepinephrine/deficiency , Adrenergic Agents/metabolism , Animals , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/metabolism , Dopamine beta-Hydroxylase/metabolism , Embryo, Mammalian , Epinephrine/metabolism , Epinephrine/pharmacology , Female , Heart/drug effects , Heart/embryology , Heart/innervation , Heart Diseases/embryology , Heart Diseases/genetics , Heart Diseases/metabolism , Isoproterenol/pharmacology , Maternal-Fetal Exchange/drug effects , Mice , Mice, Knockout , Norepinephrine/metabolism , Norepinephrine/pharmacology , Pregnancy , Up-Regulation/drug effects
6.
Mol Biotechnol ; 54(2): 350-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22706789

ABSTRACT

Adrenaline and noradrenaline are important neurotransmitter hormones that mediate physiological stress responses in adult mammals, and are essential for cardiovascular function during a critical period of embryonic/fetal development. In this study, we describe a novel mouse model system for identifying and characterizing adrenergic cells. Specifically, we generated a reporter mouse strain in which a nuclear-localized enhanced green fluorescent protein gene (nEGFP) was inserted into exon 1 of the gene encoding Phenylethanolamine n-methyltransferase (Pnmt), the enzyme responsible for production of adrenaline from noradrenaline. Our analysis demonstrates that this knock-in mutation effectively marks adrenergic cells in embryonic and adult mice. We see expression of nEGFP in Pnmt-expressing cells of the adrenal medulla in adult animals. We also note that nEGFP expression recapitulates the restricted expression of Pnmt in the embryonic heart. Finally, we show that nEGFP and Pnmt expressions are each induced in parallel during the in vitro differentiation of pluripotent mouse embryonic stem cells into beating cardiomyocytes. Thus, this new mouse genetic model should be useful for the identification and functional characterization of adrenergic cells in vitro and in vivo.


Subject(s)
Adrenal Medulla/metabolism , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Adrenal Medulla/cytology , Animals , Embryonic Stem Cells/metabolism , Epinephrine/genetics , Epinephrine/metabolism , Gene Expression , Green Fluorescent Proteins/metabolism , Mice , Mutation , Myocytes, Cardiac/metabolism , Norepinephrine/genetics , Norepinephrine/metabolism , Phenylethanolamine N-Methyltransferase/genetics , Phenylethanolamine N-Methyltransferase/metabolism , Pluripotent Stem Cells/metabolism
7.
Physiol Genomics ; 44(19): 934-47, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22911456

ABSTRACT

Adrenergic hormones are essential for early heart development. To gain insight into understanding how these hormones influence heart development, we evaluated genomic expression changes in embryonic hearts from adrenergic-deficient and wild-type control mice. To perform this study, we used a mouse model with targeted disruption of the Dopamine ß-hydroxylase (Dbh) gene, whose product is responsible for enzymatic conversion of dopamine into norepinephrine. Embryos homozygous for the null allele (Dbh(-/-)) die from heart failure beginning as early as embryonic day 10.5 (E10.5). To assess underlying causes of heart failure, we isolated hearts from Dbh(-/-) and Dbh(+/+) embryos prior to manifestation of the phenotype and examined gene expression changes using genomic Affymetrix 430A 2.0 arrays, which enabled simultaneous evaluation of >22,000 genes. We found that only 22 expressed genes showed a significant twofold or greater change, representing ~0.1% of the total genes analyzed. More than half of these genes are associated with either metabolism (31%) or signal transduction (22%). Remarkably, several of the altered genes encode for proteins that are directly involved in retinoic acid (RA) biosynthesis and transport. Subsequent evaluation showed that RA concentrations were significantly elevated by an average of ~3-fold in adrenergic-deficient (Dbh(-/-)) embryos compared with controls, thereby suggesting that RA may be an important downstream mediator of adrenergic action during embryonic heart development.


Subject(s)
Biosynthetic Pathways/genetics , Dopamine beta-Hydroxylase/genetics , Gene Expression Regulation, Developmental/physiology , Heart Failure/genetics , Heart/embryology , Tretinoin/metabolism , Animals , Dopamine beta-Hydroxylase/deficiency , Echocardiography , Gene Expression Regulation, Developmental/genetics , Heart Failure/enzymology , Mice , Mice, Knockout , Microarray Analysis , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...