Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.501
Filter
1.
PLoS Genet ; 20(4): e1011228, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38598567

ABSTRACT

The laboratory mouse has served as the premier animal model system for both basic and preclinical investigations for over a century. However, laboratory mice capture only a subset of the genetic variation found in wild mouse populations, ultimately limiting the potential of classical inbred strains to uncover phenotype-associated variants and pathways. Wild mouse populations are reservoirs of genetic diversity that could facilitate the discovery of new functional and disease-associated alleles, but the scarcity of commercially available, well-characterized wild mouse strains limits their broader adoption in biomedical research. To overcome this barrier, we have recently developed, sequenced, and phenotyped a set of 11 inbred strains derived from wild-caught Mus musculus domesticus. Each of these "Nachman strains" immortalizes a unique wild haplotype sampled from one of five environmentally distinct locations across North and South America. Whole genome sequence analysis reveals that each strain carries between 4.73-6.54 million single nucleotide differences relative to the GRCm39 mouse reference, with 42.5% of variants in the Nachman strain genomes absent from current classical inbred mouse strain panels. We phenotyped the Nachman strains on a customized pipeline to assess the scope of disease-relevant neurobehavioral, biochemical, physiological, metabolic, and morphological trait variation. The Nachman strains exhibit significant inter-strain variation in >90% of 1119 surveyed traits and expand the range of phenotypic diversity captured in classical inbred strain panels. These novel wild-derived inbred mouse strain resources are set to empower new discoveries in both basic and preclinical research.


Subject(s)
Genetic Variation , Mice, Inbred Strains , Phenotype , Animals , Mice , Mice, Inbred Strains/genetics , Genomics/methods , Animals, Wild/genetics , Genome/genetics , Polymorphism, Single Nucleotide , Haplotypes , Whole Genome Sequencing
3.
J Virol ; 97(5): e0193022, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37093008

ABSTRACT

Inbred mouse lines vary in their ability to mount protective antiretroviral immune responses, and even closely related strains can exhibit opposing phenotypes upon retroviral infection. Here, we found that 129S mice inherit a previously unknown mechanism for the production of anti-murine leukemia virus (MLV) antibodies and control of infection. The resistant phenotype in 129S1 mice is controlled by two dominant loci that are independent from known MLV resistance genes. We also show that production of anti-MLV antibodies in 129S7 mice, but not 129S1 mice, is independent of interferon gamma signaling. Thus, our data indicate that 129S mice inherit an unknown mechanism for control of MLV infection and demonstrate that there is genetic variability in 129S substrains that affects their ability to mount antiviral immune responses. IMPORTANCE Understanding the genetic basis for production of protective antiviral immune responses is crucial for the development of novel vaccines and adjuvants. Additionally, characterizing the genetic and phenotypic variability in inbred mice has implications for the selection of strains for targeted mutagenesis, choice of controls, and for broader understanding of the requirements for protective immunity.


Subject(s)
Mice, Inbred Strains , Retroviridae Infections , Animals , Mice , Immunity , Interferon-gamma , Leukemia Virus, Murine/genetics , Mice, Inbred Strains/genetics , Mice, Inbred Strains/immunology , Retroviridae Infections/immunology
4.
mBio ; 13(1): e0340021, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35089087

ABSTRACT

Host genetic determinants that underpin variation in susceptibility to systemic fungal infection are poorly understood. Genes responsible for complex traits can be identified by correlating variation in phenotype with allele in founder strains of wild mice with known genetic variation, assembled in genetic reference panels. In this work, we describe wide natural variation in both primary and acquired resistance to experimental pulmonary blastomycosis in eight founder strains, including 129, A/J, BL/6, CAST, NOD, NZO, PWK, and WSB of the Collaborative Cross collection, and the inbred DBA strain. These differences in susceptibility across strains were accompanied by sharp differences in the accumulation and function of immune cells in the lungs. Immune perturbations were mapped by identifying reagents that phenotypically mark immune cell populations in the distinct strains of mice. In particular, we uncovered marked differences between BL/6 and DBA/2 mouse strains in the development of acquired resistance. Our findings highlight the potential value in using genetic reference panels of mice, and particularly the BXD (recombinant inbred strains of mice from a cross of C57BL/6J and DBA/2J mice) collection harboring a cross between resistant BL/6 and susceptible DBA/2 mice, for unveiling genes linked with host resistance to fungal infection. IMPORTANCE Host genetic variation significantly impacts vulnerability to infectious diseases. While host variation in susceptibility to fungal infection with dimorphic fungi has long been recognized, genes that underpin this variation are poorly understood. We used a collection of seven mouse strains that represent nearly 90% of the genetic variation in mice to identify genetic variability among the strains in resistance to pulmonary infection with the dimorphic fungus Blastomyces dermatitidis. We analyzed differences between the strains in innate resistance by infecting naive mice and in acquired resistance by infecting vaccinated mice. We identified extreme variations in both innate and acquired resistance among the strains. In particular, we found sharp differences between C57BL/6 and DBA/2 strains in the ability to acquire vaccine-induced resistance. We also identified commercial reagents that allowed the phenotyping of immune cells from this strain collection of mice. Because there are additional mice harboring a genetic cross of the C57BL/6 and DBA/2 strains (BXD collection), such mice will permit future investigations to identify the genes that underlie differences in the ability to acquire resistance to infection.


Subject(s)
Blastomyces , Immunophenotyping , Mice, Inbred Strains , Animals , Mice , Blastomyces/genetics , Blastomyces/immunology , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Mice, Inbred Strains/genetics , Mice, Inbred Strains/immunology
5.
Mamm Genome ; 33(1): 31-43, 2022 03.
Article in English | MEDLINE | ID: mdl-34782917

ABSTRACT

Laboratory mouse strains have mosaic genomes derived from at least three major subspecies that are distributed in Eurasia. Here, we describe genomic variations in ten inbred strains: Mus musculus musculus-derived BLG2/Ms, NJL/Ms, CHD/Ms, SWN/Ms, and KJR/Ms; M. m. domesticus-derived PGN2/Ms and BFM/Ms; M. m. castaneus-derived HMI/Ms; and JF1/Ms and MSM/Ms, which were derived from a hybrid between M. m. musculus and M. m. castaneus. These strains were established by Prof. Moriwaki in the 1980s and are collectively named the "Mishima Battery". These strains show large phenotypic variations in body size and in many physiological traits. We resequenced the genomes of the Mishima Battery strains and performed a comparative genomic analysis with dbSNP data. More than 81 million nucleotide coordinates were identified as variant sites due to the large genetic distances among the mouse subspecies; 8,062,070 new SNP sites were detected in this study, and these may underlie the large phenotypic diversity observed in the Mishima Battery. The new information was collected in a reconstructed genome database, termed MoG+ that includes new application software and viewers. MoG+ intuitively visualizes nucleotide variants in genes and intergenic regions, and amino acid substitutions across the three mouse subspecies. We report statistical data from the resequencing and comparative genomic analyses and newly collected phenotype data of the Mishima Battery, and provide a brief description of the functions of MoG+, which provides a searchable and unique data resource of the numerous genomic variations across the three mouse subspecies. The data in MoG+ will be invaluable for research into phenotype-genotype links in diverse mouse strains.


Subject(s)
Databases, Genetic , Genome , Mice, Inbred Strains , Animals , Biomedical Research , Genomics , Mice , Mice, Inbred Strains/genetics , Nucleotides
6.
Biomolecules ; 11(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34944406

ABSTRACT

BXD recombinant inbred (RI) lines represent a genetic reference population derived from a cross between C57BL/6J mice (B6) and DBA/2J mice (D2), which through meiotic recombination events possesses recombinant chromosomes containing B6 or D2 haplotype segments. The quantitative trait loci (QTLs) are the locations of segregating genetic polymorphisms and are fundamental to understanding genetic diversity in human disease susceptibility and severity. QTL mapping represents the typical approach for identifying naturally occurring polymorphisms that influence complex phenotypes. In this process, genotypic values at markers of known genomic locations are associated with phenotypic values measured in a segregating population. Indeed, BXD RI strains provide a powerful tool to study neurotoxicity induced by different substances. In this review, we describe the use of BXD RI lines to understand the underlying mechanisms of neurotoxicity in response to ethanol and cocaine, as well as metals and pesticide exposures.


Subject(s)
Mice, Inbred Strains/genetics , Neurotoxicity Syndromes/genetics , Quantitative Trait Loci , Animals , Chromosome Mapping , Disease Models, Animal , Haplotypes , Male , Mice , Neurotoxicity Syndromes/etiology , Recombination, Genetic
7.
Food Funct ; 12(22): 11077-11105, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34672309

ABSTRACT

Mouse models are an essential tool in different areas of research, including nutrition and phytochemical research. Traditional inbred mouse models have allowed the discovery of therapeutical targets and mechanisms of action and expanded our knowledge of health and disease. However, these models lack the genetic variability typically found in human populations, which hinders the translatability of the results found in mice to humans. The development of genetically diverse mouse models, such as the collaborative cross (CC) or the diversity outbred (DO) models, has been a useful tool to overcome this obstacle in many fields, such as cancer, immunology and toxicology. However, these tools have not yet been widely adopted in the field of phytochemical research. As demonstrated in other disciplines, use of CC and DO models has the potential to provide invaluable insights for translation of phytochemicals from rodents to humans, which are desperately needed given the challenges and numerous failed clinical trials in this field. These models may prove informative for personalized use of phytochemicals in humans, including: predicting interindividual variability in phytochemical bioavailability and efficacy, identifying genetic loci or genes governing response to phytochemicals, identifying phytochemical mechanisms of action and therapeutic targets, and understanding the impact of genetic variability on individual response to phytochemicals. Such insights would prove invaluable for personalized implementation of phytochemicals in humans. This review will focus on the current work performed with genetically diverse mouse populations, and the research opportunities and advantages that these models can offer to phytochemical research.


Subject(s)
Disease Models, Animal , Genetic Variation/genetics , Mice, Inbred Strains/genetics , Nutritional Physiological Phenomena , Phytochemicals , Animals , Drug Evaluation, Preclinical , Humans , Mice , Nutritional Physiological Phenomena/drug effects , Nutritional Physiological Phenomena/genetics , Phytochemicals/administration & dosage , Phytochemicals/pharmacology , Translational Research, Biomedical
8.
Immunol Lett ; 237: 3-10, 2021 09.
Article in English | MEDLINE | ID: mdl-34174253

ABSTRACT

Many studies of the autoimmune disease Sjögren's syndrome have been performed using spontaneous mouse models. In the present study, we describe the characteristics of McH/lpr-RA1 mice and propose their use as a novel murine model of autoimmune sialadenitis. The McH/lpr-RA1 mouse is a recombinant congenic strain derived from generation F54 or more of MRL-Faslpr x (MRL- Faslpr x C3H- Faslpr) F1. We show for the first time that this mouse spontaneously develops autoimmune sialadenitis and vasculitis in submandibular gland tissues. Sialadenitis was accompanied by extensive inflammatory cell infiltration and tissue destruction. Immunohistochemical studies revealed that the salivary gland lesions strongly expressed four sialadenitis-related molecules: SSA and SSB (autoantigens of Sjögren's syndrome), gp91phox (an accelerator of reactive oxygen species production) and single strand DNA (a marker of apoptotic cells). In contrast, expression of aquaporin-5 (AQP5), which stimulates salivary secretion was weak or negligible. Statistical correlation analyses indicated that the apoptosis of salivary gland cells provoked by oxidative stress contributed to the severe sialadenitis and reduced expression of AQP5. Our study has demonstrated that McH/lpr-RA1 mice spontaneously develop the pathognomonic features of autoimmune sialadenitis and thus could be used as a new animal model of Sjögren's syndrome.


Subject(s)
Autoimmune Diseases/immunology , Disease Models, Animal , Mice, Inbred Strains/immunology , Mice, Mutant Strains/immunology , Sialadenitis/immunology , Sjogren's Syndrome , Vasculitis/immunology , Animals , Animals, Congenic , Apoptosis , Aquaporin 5/biosynthesis , Aquaporin 5/genetics , Autoantigens/biosynthesis , Autoantigens/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , DNA, Single-Stranded/analysis , Female , Genetic Predisposition to Disease , Mice , Mice, Inbred C3H , Mice, Inbred Strains/genetics , Mice, Mutant Strains/genetics , NADPH Oxidase 2/biosynthesis , NADPH Oxidase 2/genetics , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/genetics , Severity of Illness Index , Sialadenitis/genetics , Sialadenitis/pathology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology , Submandibular Gland/metabolism , Submandibular Gland/pathology , Vasculitis/genetics , Vasculitis/pathology , SS-B Antigen
9.
Exp Anim ; 70(2): 145-160, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33441510

ABSTRACT

The inbred mouse strain C57BL/6 has been widely used as a background strain for spontaneous and induced mutations. Developed in the 1930s, the C57BL/6 strain diverged into two major groups in the 1950s, namely, C57BL/6J and C57BL/6N, and more than 20 substrains have been established from them worldwide. We previously reported genetic differences among C57BL/6 substrains in 2009 and 2015. Since then, dozens of reports have been published on phenotypic differences in behavioral, neurological, cardiovascular, and metabolic traits. Substrains need to be chosen according to the purpose of the study because phenotypic differences might affect the experimental results. In this paper, we review recent reports of phenotypic and genetic differences among C57BL/6 substrains, focus our attention on the proper use of C57BL/6 and other inbred strains in the era of genome editing, and provide the life science research community wider knowledge about this subject.


Subject(s)
Mice, Inbred Strains/physiology , Phenotype , Animals , Mice , Mice, Inbred Strains/genetics , Species Specificity
10.
Mol Genet Metab ; 132(2): 139-145, 2021 02.
Article in English | MEDLINE | ID: mdl-33483254

ABSTRACT

The glutaric acidurias are a group of inborn errors of metabolism with different etiologies. Glutaric aciduria type 3 (GA3) is a biochemical phenotype with uncertain clinical relevance caused by a deficiency of succinyl-CoA:glutarate-CoA transferase (SUGCT). SUGCT catalyzes the succinyl-CoA-dependent conversion of glutaric acid into glutaryl-CoA preventing urinary loss of the organic acid. Here, we describe the presence of a GA3 trait in mice of 129 substrains due to SUGCT deficiency, which was identified by screening of urine organic acid profiles obtained from different inbred mouse strains including 129S2/SvPasCrl. Molecular and biochemical analyses in an F2 population of the parental C57BL/6J and 129S2/SvPasCrl strains (B6129F2) confirmed that the GA3 trait occurred in Sugct129/129 animals. We evaluated the impact of SUGCT deficiency on metabolite accumulation in the glutaric aciduria type 1 (GA1) mouse model. We found that GA1 mice with SUGCT deficiency have decreased excretion of urine 3-hydroxyglutaric acid and decreased levels glutarylcarnitine in urine, plasma and kidney. Our work demonstrates that SUGCT contributes to the production of glutaryl-CoA under conditions of low and pathologically high glutaric acid levels. Our work also highlights the notion that unexpected biochemical phenotypes can occur in widely used inbred animal lines.


Subject(s)
Acyltransferases/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Metabolic Diseases/genetics , Mice, Inbred Strains/genetics , Oxidoreductases/deficiency , Transferases/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Disease Models, Animal , Glutarates/metabolism , Humans , Lysine/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phenotype
11.
Behav Genet ; 51(1): 68-81, 2021 01.
Article in English | MEDLINE | ID: mdl-32939625

ABSTRACT

We conducted whole-genome sequencing of four inbred mouse strains initially selected for high (H1, H2) or low (L1, L2) open-field activity (OFA), and then examined strain distribution patterns for all DNA variants that differed between their BALB/cJ and C57BL/6J parental strains. Next, we assessed genome-wide sharing (3,678,826 variants) both between and within the High and Low Activity strains. Results suggested that about 10% of these DNA variants may be associated with OFA, and clearly demonstrated its polygenic nature. Finally, we conducted bioinformatic analyses of functional genomics data from mouse, rat, and human to refine previously identified quantitative trait loci (QTL) for anxiety-related measures. This combination of sequence analysis and genomic-data integration facilitated refinement of previously intractable QTL findings, and identified possible genes for functional follow-up studies.


Subject(s)
Anxiety/genetics , Mice, Inbred Strains/genetics , Open Field Test/physiology , Animals , Anxiety Disorders/genetics , Chromosome Mapping/methods , Computational Biology/methods , Disease Models, Animal , Genomics/methods , Genotype , Humans , Mice , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Rats , Exome Sequencing/methods
12.
J Gerontol A Biol Sci Med Sci ; 76(4): 552-560, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33211821

ABSTRACT

Aging is associated with functional and metabolic decline and is a risk factor for all noncommunicable diseases. Even though mice are routinely used for modeling human aging and aging-related conditions, no comprehensive assessment to date has been conducted on normative mouse aging. To address this gap, the Study of Longitudinal Aging in Mice (SLAM) was designed and implemented by the National Institute on Aging (NIA/NIH) as the mouse counterpart to the Baltimore Longitudinal Study of Aging (BLSA). In this manuscript, we describe the premise, study design, methodologies, and technologies currently employed in SLAM. We also discuss current and future study directions. In this large population mouse study, inbred C57BL/6J and outbred UM-HET3 mice of both sexes are longitudinally evaluated for functional, phenotypic, and biological health, and collection of biospecimens is conducted throughout their life span. Within the longitudinal cohorts, a cross-sectional arm of the study has also been implemented for the well-controlled collection of tissues to generate a biorepository. SLAM and studies stemming from SLAM seek to identify and characterize phenotypic and biological predictors of mouse aging and age-associated conditions, examine the degrees of functional and biomolecular variability that occur within inbred and genetically heterogeneous mouse populations with age, and assess whether these changes are consistent with alterations observed in human aging in BLSA. The findings from these studies will be critical for evaluating the utility of mouse models for studying different aspects of aging, both in terms of interpreting prior findings and designing and implementing future studies.


Subject(s)
Aging/physiology , Longevity/physiology , Models, Animal , Animals , Biological Variation, Population , Biomarkers/analysis , Biotechnology/methods , Genetic Variation , Humans , Life Expectancy , Longitudinal Studies , Mice , Mice, Inbred Strains/genetics , Mice, Inbred Strains/metabolism , Physical Functional Performance , Procedures and Techniques Utilization , Research Design
13.
Radiat Res ; 194(5): 485-499, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32991727

ABSTRACT

We present a novel mathematical formalism to predict the kinetics of DNA damage repair after exposure to both low- and high-LET radiation (X rays; 350 MeV/n 40Ar; 600 MeV/n 56Fe). Our method is based on monitoring DNA damage repair protein 53BP1 that forms radiation-induced foci (RIF) at locations of DNA double-strand breaks (DSB) in the nucleus and comparing its expression in primary skin fibroblasts isolated from 15 mice strains. We previously reported strong evidence for clustering of nearby DSB into single repair units as opposed to the classic "contact-first" model where DSB are considered immobile. Here we apply this clustering model to evaluate the number of remaining RIF over time. We also show that the newly introduced kinetic metrics can be used as surrogate biomarkers for in vivo radiation toxicity, with potential applications in radiotherapy and human space exploration. In particular, we observed an association between the characteristic time constant of RIF repair measured in vitro and survival levels of immune cells collected from irradiated mice. Moreover, the speed of DNA damage repair correlated not only with radiation-induced cellular survival in vivo, but also with spontaneous cancer incidence data collected from the Mouse Tumor Biology database, suggesting a relationship between the efficiency of DSB repair after irradiation and cancer risk.


Subject(s)
DNA Repair , DNA/radiation effects , Mice, Inbred Strains/genetics , Radiation Tolerance/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Aerospace Medicine , Animals , Cells, Cultured , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Damage , Female , Fibroblasts/radiation effects , Heavy Ions , Incidence , Kinetics , Linear Energy Transfer , Male , Mice , Models, Genetic , Neoplasms/epidemiology , Neoplasms/genetics , Neoplasms/veterinary , Radiation Exposure , Relative Biological Effectiveness , Risk , Rodent Diseases/epidemiology , Rodent Diseases/genetics
14.
PLoS One ; 15(9): e0238893, 2020.
Article in English | MEDLINE | ID: mdl-32956361

ABSTRACT

Utilization of murine models remains a valuable tool in biomedical research, yet, disease phenotype of mice across studies can vary considerably. With advances in next generation sequencing, it is increasingly recognized that inconsistencies in host phenotype can be attributed, at least in part, to differences in gut bacterial composition. Research with inbred murine strains demonstrates that housing conditions play a significant role in variations of gut bacterial composition, however, few studies have assessed whether observed variation influences host phenotype in response to an intervention. Our study initially sought to examine the effects of a long-term (9-months) dietary intervention (i.e., diets with distinct fatty acid compositions) on the metabolic health, in particular glucose homeostasis, of genetically-outbred male and female CD-1 mice. Yet, mice were shipped from two different husbandry facilities of the same commercial vendor (Cohort A and B, respectively), and we observed throughout the study that diet, sex, and aging differentially influenced the metabolic phenotype of mice depending on their husbandry facility of origin. Examination of the colonic bacteria of mice revealed distinct bacterial compositions, including 23 differentially abundant genera and an enhanced alpha diversity in mice of Cohort B compared to Cohort A. We also observed that a distinct metabolic phenotype was linked with these differentially abundant bacteria and indices of alpha diversity. Our findings support that metabolic phenotypic variation of mice of the same strain but shipped from different husbandry facilities may be influenced by their colonic bacterial community structure. Our work is an important precautionary note for future research of metabolic diseases via mouse models, particularly those that seek to examine factors such diet, sex, and aging.


Subject(s)
Bacteria/classification , Diet/adverse effects , Feces/microbiology , Glucose/metabolism , High-Throughput Nucleotide Sequencing/methods , Mice, Inbred Strains/genetics , Animal Husbandry , Animals , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Female , Gastrointestinal Microbiome/drug effects , Male , Mice , Models, Animal , Phenotype , Phylogeny , Sequence Analysis, DNA
15.
J Mol Med (Berl) ; 98(9): 1215-1234, 2020 09.
Article in English | MEDLINE | ID: mdl-32712726

ABSTRACT

The laboratory mouse is the most commonly used mammalian model for biomedical research. An enormous number of mouse models, such as gene knockout, knockin, and overexpression transgenic mice, have been created over the years. A common practice to maintain a genetically modified mouse line is backcrossing with standard inbred mice over several generations. However, the choice of inbred mouse for backcrossing is critical to phenotypic characterization because phenotypic variabilities are often observed between mice with different genetic backgrounds. In this review, the major features of commonly used inbred mouse lines are discussed. The aim is to provide information for appropriate selection of inbred mouse lines for genetic and behavioral studies.


Subject(s)
Mice, Inbred Strains , Phenotype , Animals , Breeding , Disease Models, Animal , Genetic Background , Mice , Mice, Inbred Strains/genetics , Mice, Knockout , Mice, Transgenic , Models, Animal
16.
Genes Genomics ; 42(9): 1023-1033, 2020 09.
Article in English | MEDLINE | ID: mdl-32712838

ABSTRACT

BACKGROUND: p19arf, primarily known as a tumor suppressor, has also been reported to play an essential role in normal development of mouse eyes. Consistently, lack of p19arf has been associated with ocular defects, but the mixed background of the knockout (KO) mouse strain used raised a concern on the accuracy of the phenotypes observed in association with the targeted gene due to genetic heterogeneity. OBJECT: We carried out a study to investigate into the effect of genetic background on the manifestation of p19arf KO associated phenotypes. METHODS: We characterized the phenotypes of novel p19arf KO mouse lines generated in FVB/N and C57BL/6J using a transcription activator-like effector nuclease (TALEN) system in comparison to the reported phenotypes of three other p19arf-deficient mouse lines generated using homologous recombination. RESULTS: Ninety-five percent of FVB/N-p19arf KO mice showed ocular opacity from week 4 after birth which worsened rapidly until week 6, while such abnormality was absent in C57BL/6J-p19arf KO mice up to the age of 26 weeks. Histopathological analysis revealed retrolental masses and dysplasia in the retinal layer in FVB/N-p19arf KO mice from week 4. Besides these, both strains developed normally from birth to week 26 without increased tumorigenesis except for a subcutaneous tumor found in a C57BL/6J-p19arf KO mouse. CONCLUSION: Our findings demonstrated surprisingly variable manifestation of p19arf-linked phenotypes between FVB/N and C57BL/6J mice, and furthermore between our mouse lines and the established lines, indicating a critical impact of genetic background on functional study of genes using gene targeting strategies in mice.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Mice, Inbred Strains/genetics , Transcription Activator-Like Effector Nucleases/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Eye/embryology , Eye/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ocular Physiological Phenomena/genetics , Phenotype , Transcription Activator-Like Effector Nucleases/physiology , Transcription Activator-Like Effectors/genetics , Vision, Ocular/genetics , Vision, Ocular/physiology
17.
Mol Metab ; 40: 101027, 2020 10.
Article in English | MEDLINE | ID: mdl-32480041

ABSTRACT

OBJECTIVES: Apoptosis-Inducing Factor (AIF) is a protein involved in mitochondrial electron transport chain assembly/stability and programmed cell death. The relevant role of this protein is underlined because mutations altering mitochondrial AIF properties result in acute pediatric mitochondriopathies and tumor metastasis. By generating an original AIF-deficient mouse strain, this study attempted to analyze, in a single paradigm, the cellular and developmental metabolic consequences of AIF loss and the subsequent oxidative phosphorylation (OXPHOS) dysfunction. METHODS: We developed a novel AIF-deficient mouse strain and assessed, using molecular and cell biology approaches, the cellular, embryonic, and adult mice phenotypic alterations. Additionally, we conducted ex vivo assays with primary and immortalized AIF knockout mouse embryonic fibroblasts (MEFs) to establish the cell death characteristics and the metabolic adaptive responses provoked by the mitochondrial electron transport chain (ETC) breakdown. RESULTS: AIF deficiency destabilized mitochondrial ETC and provoked supercomplex disorganization, mitochondrial transmembrane potential loss, and high generation of mitochondrial reactive oxygen species (ROS). AIF-/Y MEFs counterbalanced these OXPHOS alterations by mitochondrial network reorganization and a metabolic reprogramming toward anaerobic glycolysis illustrated by the AMPK phosphorylation at Thr172, the overexpression of the glucose transporter GLUT-4, the subsequent enhancement of glucose uptake, and the anaerobic lactate generation. A late phenotype was characterized by the activation of P53/P21-mediated senescence. Notably, approximately 2% of AIF-/Y MEFs diminished both mitochondrial mass and ROS levels and spontaneously proliferated. These cycling AIF-/Y MEFs were resistant to caspase-independent cell death inducers. The AIF-deficient mouse strain was embryonic lethal between E11.5 and E13.5 with energy loss, proliferation arrest, and increased apoptotic levels. Contrary to AIF-/Y MEFs, the AIF KO embryos were unable to reprogram their metabolism toward anaerobic glycolysis. Heterozygous AIF+/- females displayed progressive bone marrow, thymus, and spleen cellular loss. In addition, approximately 10% of AIF+/- females developed perinatal hydrocephaly characterized by brain development impairment, meningeal fibrosis, and medullar hemorrhages; those mice died 5 weeks after birth. AIF+/- with hydrocephaly exhibited loss of ciliated epithelium in the ependymal layer. This phenotype was triggered by the ROS excess. Accordingly, it was possible to diminish the occurrence of hydrocephalus AIF+/- females by supplying dams and newborns with an antioxidant in drinking water. CONCLUSIONS: In a single knockout model and at 3 different levels (cell, embryo, and adult mice) we demonstrated that by controlling the mitochondrial OXPHOS/metabolism, AIF is a key factor regulating cell differentiation and fate. Additionally, by providing new insights into the pathological consequences of mitochondrial OXPHOS dysfunction, our new findings pave the way for novel pharmacological strategies.


Subject(s)
Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Animals , Apoptosis/physiology , Caspases/metabolism , Cell Respiration , Female , Fibroblasts/metabolism , Genetic Engineering/methods , Glycolysis/genetics , Hydrocephalus/metabolism , Male , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains/genetics , Mitochondria/metabolism , Models, Animal , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism
18.
Mol Neurobiol ; 57(5): 2301-2313, 2020 May.
Article in English | MEDLINE | ID: mdl-32020500

ABSTRACT

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition with unknown etiology. Recent experimental evidences suggest the contribution of non-coding RNAs (ncRNAs) in the pathophysiology of ASD. In this work, we aimed to investigate the expression profile of the ncRNA class of circular RNAs (circRNAs) in the hippocampus of the BTBR T + tf/J (BTBR) mouse model and age-matched C57BL/6J (B6) mice. Alongside, we analyzed BTBR hippocampal gene expression profile to evaluate possible correlations between the differential abundance of circular and linear gene products. From RNA sequencing data, we identified circRNAs highly modulated in BTBR mice. Thirteen circRNAs and their corresponding linear isoforms were validated by RT-qPCR analysis. The BTBR-regulated circCdh9 was better characterized in terms of molecular structure and expression, highlighting altered levels not only in the hippocampus, but also in the cerebellum, prefrontal cortex, and amygdala. Finally, gene expression analysis of the BTBR hippocampus pinpointed altered biological and molecular pathways relevant for the ASD phenotype. By comparison of circRNA and gene expression profiles, we identified 6 genes significantly regulated at either circRNA or mRNA gene products, suggesting low overall correlation between circRNA and host gene expression. In conclusion, our results indicate a consistent deregulation of circRNA expression in the hippocampus of BTBR mice. ASD-related circRNAs should be considered in functional studies to identify their contribution to the etiology of the disorder. In addition, as abundant and highly stable molecules, circRNAs represent interesting potential biomarkers for autism.


Subject(s)
Autism Spectrum Disorder/metabolism , Disease Models, Animal , Hippocampus/metabolism , Mice, Inbred Strains/metabolism , Mice, Mutant Strains/metabolism , RNA, Circular/biosynthesis , RNA, Messenger/biosynthesis , Animals , Autism Spectrum Disorder/genetics , Brain Chemistry , Gene Expression Profiling , Gene Ontology , Humans , Male , Mice, Inbred C57BL , Mice, Inbred Strains/genetics , Mice, Mutant Strains/genetics , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
19.
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
20.
Osteoarthritis Cartilage ; 28(4): 516-527, 2020 04.
Article in English | MEDLINE | ID: mdl-31945456

ABSTRACT

OBJECTIVE: To investigate the transcriptomic differences in chondrocytes obtained from LG/J (large, healer) and SM/J (small, non-healer) murine strains in an attempt to discern the molecular pathways implicated in cartilage regeneration and susceptibility to osteoarthritis (OA). DESIGN: We performed RNA-sequencing on chondrocytes derived from LG/J (n = 16) and SM/J (n = 16) mice. We validated the expression of candidate genes and compared single nucleotide polymorphisms (SNPs) between the two mouse strains. We also examined gene expression of positional candidates for ear pinna regeneration and long bone length quantitative trait loci (QTLs) that display differences in cartilaginous expression. RESULTS: We observed a distinct genetic heterogeneity between cells derived from LG/J and SM/J mouse strains. We found that gene ontologies representing cell development, cartilage condensation, and regulation of cell differentiation were enriched in LG/J chondrocytes. In contrast, gene ontologies enriched in the SM/J chondrocytes were mainly related to inflammation and degeneration. Moreover, SNP analysis revealed that multiple validated genes vary in sequence between LG/J and SM/J in coding and highly conserved noncoding regions. Finally, we showed that most QTLs have 20-30% of their positional candidates displaying differential expression between the two mouse strains. CONCLUSIONS: While the enrichment of pathways related to cell differentiation, cartilage development and cartilage condensation infers superior healing potential of LG/J strain, the enrichment of pathways related to cytokine production, immune cell activation and inflammation entails greater susceptibility of SM/J strain to OA. These data provide novel insights into chondrocyte transcriptome and aid in identification of the quantitative trait genes and molecular differences underlying the phenotypic differences associated with individual QTLs.


Subject(s)
Cartilage/physiology , Chondrocytes/metabolism , Osteoarthritis/genetics , Regeneration/genetics , Animals , Carbonic Anhydrase II/genetics , Cartilage, Articular/physiology , Ear Auricle , Ear Cartilage/physiology , Gene Expression Profiling , Genetic Predisposition to Disease , Mice , Mice, Inbred Strains/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , RNA-Seq , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...