Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Genet Med ; 25(7): 100835, 2023 Jul.
Article En | MEDLINE | ID: mdl-36999555

PURPOSE: Miller-Dieker syndrome is caused by a multiple gene deletion, including PAFAH1B1 and YWHAE. Although deletion of PAFAH1B1 causes lissencephaly unambiguously, deletion of YWHAE alone has not clearly been linked to a human disorder. METHODS: Cases with YWHAE variants were collected through international data sharing networks. To address the specific impact of YWHAE loss of function, we phenotyped a mouse knockout of Ywhae. RESULTS: We report a series of 10 individuals with heterozygous loss-of-function YWHAE variants (3 single-nucleotide variants and 7 deletions <1 Mb encompassing YWHAE but not PAFAH1B1), including 8 new cases and 2 follow-ups, added with 5 cases (copy number variants) from literature review. Although, until now, only 1 intragenic deletion has been described in YWHAE, we report 4 new variants specifically in YWHAE (3 splice variants and 1 intragenic deletion). The most frequent manifestations are developmental delay, delayed speech, seizures, and brain malformations, including corpus callosum hypoplasia, delayed myelination, and ventricular dilatation. Individuals with variants affecting YWHAE alone have milder features than those with larger deletions. Neuroanatomical studies in Ywhae-/- mice revealed brain structural defects, including thin cerebral cortex, corpus callosum dysgenesis, and hydrocephalus paralleling those seen in humans. CONCLUSION: This study further demonstrates that YWHAE loss-of-function variants cause a neurodevelopmental disease with brain abnormalities.


Classical Lissencephalies and Subcortical Band Heterotopias , Intellectual Disability , Lissencephaly , Neurodevelopmental Disorders , Humans , Animals , Mice , Brain/abnormalities , Lissencephaly/genetics , Intellectual Disability/genetics , 14-3-3 Proteins/genetics
2.
Int J Mol Sci ; 23(19)2022 Sep 29.
Article En | MEDLINE | ID: mdl-36232804

CHARGE syndrome is a rare congenital disorder frequently caused by mutations in the chromodomain helicase DNA-binding protein-7 CHD7. Here, we developed and systematically characterized two genetic mouse models with identical, heterozygous loss-of-function mutation of the Chd7 gene engineered on inbred and outbred genetic backgrounds. We found that both models showed consistent phenotypes with the core clinical manifestations seen in CHARGE syndrome, but the phenotypes in the inbred Chd7 model were more severe, sometimes having reduced penetrance and included dysgenesis of the corpus callosum, hypoplasia of the hippocampus, abnormal retrosplenial granular cortex, ventriculomegaly, hyperactivity, growth delays, impaired grip strength and repetitive behaviors. Interestingly, we also identified previously unreported features including reduced levels of basal insulin and reduced blood lipids. We suggest that the phenotypic variation reported in individuals diagnosed with CHARGE syndrome is likely due to the genetic background and modifiers. Finally, our study provides a valuable resource, making it possible for mouse biologists interested in Chd7 to make informed choices on which mouse model they should use to study phenotypes of interest and investigate in more depth the underlying cellular and molecular mechanisms.


CHARGE Syndrome , DNA-Binding Proteins/metabolism , Animals , CHARGE Syndrome/diagnosis , CHARGE Syndrome/genetics , Corpus Callosum/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , Insulins/genetics , Mice , Mutation
3.
Biol Psychiatry ; 92(4): 323-334, 2022 08 15.
Article En | MEDLINE | ID: mdl-35227461

BACKGROUND: The discovery of coding variants in genes that confer risk of intellectual disability (ID) is an important step toward understanding the pathophysiology of this common developmental disability. METHODS: Homozygosity mapping, whole-exome sequencing, and cosegregation analyses were used to identify gene variants responsible for syndromic ID with autistic features in two independent consanguineous families from the Arabian Peninsula. For in vivo functional studies of the implicated gene's function in cognition, Drosophila melanogaster and mice with targeted interference of the orthologous gene were used. Behavioral, electrophysiological, and structural magnetic resonance imaging analyses were conducted for phenotypic testing. RESULTS: Homozygous premature termination codons in PDZD8, encoding an endoplasmic reticulum-anchored lipid transfer protein, showed cosegregation with syndromic ID in both families. Drosophila melanogaster with knockdown of the PDZD8 ortholog exhibited impaired long-term courtship-based memory. Mice homozygous for a premature termination codon in Pdzd8 exhibited brain structural, hippocampal spatial memory, and synaptic plasticity deficits. CONCLUSIONS: These data demonstrate the involvement of homozygous loss-of-function mutations in PDZD8 in a neurodevelopmental cognitive disorder. Model organisms with manipulation of the orthologous gene replicate aspects of the human phenotype and suggest plausible pathophysiological mechanisms centered on disrupted brain development and synaptic function. These findings are thus consistent with accruing evidence that synaptic defects are a common denominator of ID and other neurodevelopmental conditions.


Cognitive Dysfunction , Intellectual Disability , Adaptor Proteins, Signal Transducing/genetics , Animals , Cognitive Dysfunction/genetics , Consanguinity , Drosophila , Drosophila melanogaster , Humans , Intellectual Disability/genetics , Mice , Mutation/genetics
4.
PLoS Genet ; 18(3): e1010114, 2022 03.
Article En | MEDLINE | ID: mdl-35298461

The highly evolutionarily conserved transport protein particle (TRAPP) complexes (TRAPP II and III) perform fundamental roles in subcellular trafficking pathways. Here we identified biallelic variants in TRAPPC10, a component of the TRAPP II complex, in individuals with a severe microcephalic neurodevelopmental disorder. Molecular studies revealed a weakened interaction between mutant TRAPPC10 and its putative adaptor protein TRAPPC2L. Studies of patient lymphoblastoid cells revealed an absence of TRAPPC10 alongside a concomitant absence of TRAPPC9, another key TRAPP II complex component associated with a clinically overlapping neurodevelopmental disorder. The TRAPPC9/10 reduction phenotype was recapitulated in TRAPPC10-/- knockout cells, which also displayed a membrane trafficking defect. Notably, both the reduction in TRAPPC9 levels and the trafficking defect in these cells could be rescued by wild type but not mutant TRAPPC10 gene constructs. Moreover, studies of Trappc10-/- knockout mice revealed neuroanatomical brain defects and microcephaly, paralleling findings seen in the human condition as well as in a Trappc9-/- mouse model. Together these studies confirm autosomal recessive TRAPPC10 variants as a cause of human disease and define TRAPP-mediated pathomolecular outcomes of importance to TRAPPC9 and TRAPPC10 mediated neurodevelopmental disorders in humans and mice.


Microcephaly , Neurodevelopmental Disorders , Animals , Humans , Mice , Microcephaly/genetics , Neurodevelopmental Disorders/genetics , Phenotype
5.
Mol Biol Evol ; 38(12): 5655-5663, 2021 12 09.
Article En | MEDLINE | ID: mdl-34464968

A nonsense allele at rs1343879 in human MAGEE2 on chromosome X has previously been reported as a strong candidate for positive selection in East Asia. This premature stop codon causing ∼80% protein truncation is characterized by a striking geographical pattern of high population differentiation: common in Asia and the Americas (up to 84% in the 1000 Genomes Project East Asians) but rare elsewhere. Here, we generated a Magee2 mouse knockout mimicking the human loss-of-function mutation to study its functional consequences. The Magee2 null mice did not exhibit gross abnormalities apart from enlarged brain structures (13% increased total brain area, P = 0.0022) in hemizygous males. The area of the granular retrosplenial cortex responsible for memory, navigation, and spatial information processing was the most severely affected, exhibiting an enlargement of 34% (P = 3.4×10-6). The brain size in homozygous females showed the opposite trend of reduced brain size, although this did not reach statistical significance. With these insights, we performed human association analyses between brain size measurements and rs1343879 genotypes in 141 Chinese volunteers with brain MRI scans, replicating the sexual dimorphism seen in the knockout mouse model. The derived stop gain allele was significantly associated with a larger volume of gray matter in males (P = 0.00094), and smaller volumes of gray (P = 0.00021) and white (P = 0.0015) matter in females. It is unclear whether or not the observed neuroanatomical phenotypes affect behavior or cognition, but it might have been the driving force underlying the positive selection in humans.


Antigens, Neoplasm/metabolism , Brain , Proteins/metabolism , Sex Characteristics , Alleles , Animals , Brain/diagnostic imaging , Female , Humans , Male , Mice , Mice, Knockout , Organ Size , Phenotype
7.
Nat Commun ; 12(1): 467, 2021 01 20.
Article En | MEDLINE | ID: mdl-33473114

Osteoarthritis causes debilitating pain and disability, resulting in a considerable socioeconomic burden, yet no drugs are available that prevent disease onset or progression. Here, we develop, validate and use rapid-throughput imaging techniques to identify abnormal joint phenotypes in randomly selected mutant mice generated by the International Knockout Mouse Consortium. We identify 14 genes with functional involvement in osteoarthritis pathogenesis, including the homeobox gene Pitx1, and functionally characterize 6 candidate human osteoarthritis genes in mouse models. We demonstrate sensitivity of the methods by identifying age-related degenerative joint damage in wild-type mice. Finally, we phenotype previously generated mutant mice with an osteoarthritis-associated polymorphism in the Dio2 gene by CRISPR/Cas9 genome editing and demonstrate a protective role in disease onset with public health implications. We hope this expanding resource of mutant mice will accelerate functional gene discovery in osteoarthritis and offer drug discovery opportunities for this common, incapacitating chronic disease.


Genetic Association Studies , Genetic Predisposition to Disease/genetics , Osteoarthritis/genetics , Animals , Bone and Bones/pathology , CRISPR-Cas Systems , Cartilage/pathology , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Models, Animal , Drug Discovery , Gene Editing , Gonadotropin-Releasing Hormone/genetics , Iodide Peroxidase , Mice , Mice, Knockout , Osteoarthritis/pathology , Osteoarthritis/surgery , Paired Box Transcription Factors/genetics , Phenotype , Iodothyronine Deiodinase Type II
8.
Hum Genet ; 140(6): 885-896, 2021 Jun.
Article En | MEDLINE | ID: mdl-33417013

The 22q11.2 deletion syndrome (22q11DS) is associated with a wide spectrum of cognitive and psychiatric symptoms. Despite the considerable work performed over the past 20 years, the genetic etiology of the neurodevelopmental phenotype remains speculative. Here, we report de novo heterozygous truncating variants in the HIRA (Histone cell cycle regulation defective, S. Cerevisiae, homolog of, A) gene associated with a neurodevelopmental disorder in two unrelated patients. HIRA is located within the commonly deleted region of the 22q11DS and encodes a histone chaperone that regulates neural progenitor proliferation and neurogenesis, and that belongs to the WD40 Repeat (WDR) protein family involved in brain development and neuronal connectivity. To address the specific impact of HIRA haploinsufficiency in the neurodevelopmental phenotype of 22q11DS, we combined Hira knock-down strategies in developing mouse primary hippocampal neurons, and the direct study of brains from heterozygous Hira+/- mice. Our in vitro analyses revealed that Hira gene is mostly expressed during neuritogenesis and early dendritogenesis stages in mouse total brain and in developing primary hippocampal neurons. Moreover, shRNA knock-down experiments showed that a twofold decrease of endogenous Hira expression level resulted in an impaired dendritic growth and branching in primary developing hippocampal neuronal cultures. In parallel, in vivo analyses demonstrated that Hira+/- mice displayed subtle neuroanatomical defects including a reduced size of the hippocampus, the fornix and the corpus callosum. Our results suggest that HIRA haploinsufficiency would likely contribute to the complex pathophysiology of the neurodevelopmental phenotype of 22q11DS by impairing key processes in neurogenesis and by causing neuroanatomical defects during cerebral development.


Cell Cycle Proteins/genetics , DiGeorge Syndrome/genetics , Haploinsufficiency , Histone Chaperones/genetics , Neurodevelopmental Disorders/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Child , Child, Preschool , Corpus Callosum/metabolism , Corpus Callosum/pathology , DiGeorge Syndrome/metabolism , DiGeorge Syndrome/pathology , Female , Fornix, Brain/metabolism , Fornix, Brain/pathology , Gene Expression , Heterozygote , Hippocampus/metabolism , Hippocampus/pathology , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/deficiency , Histone Chaperones/metabolism , Humans , Mice , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurogenesis/genetics , Neurons/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/metabolism
9.
Am J Hum Genet ; 107(6): 1170-1177, 2020 12 03.
Article En | MEDLINE | ID: mdl-33232677

KDM4B is a lysine-specific demethylase with a preferential activity on H3K9 tri/di-methylation (H3K9me3/2)-modified histones. H3K9 tri/di-demethylation is an important epigenetic mechanism responsible for silencing of gene expression in animal development and cancer. However, the role of KDM4B on human development is still poorly characterized. Through international data sharing, we gathered a cohort of nine individuals with mono-allelic de novo or inherited variants in KDM4B. All individuals presented with dysmorphic features and global developmental delay (GDD) with language and motor skills most affected. Three individuals had a history of seizures, and four had anomalies on brain imaging ranging from agenesis of the corpus callosum with hydrocephalus to cystic formations, abnormal hippocampi, and polymicrogyria. In mice, lysine demethylase 4B is expressed during brain development with high levels in the hippocampus, a region important for learning and memory. To understand how KDM4B variants can lead to GDD in humans, we assessed the effect of KDM4B disruption on brain anatomy and behavior through an in vivo heterozygous mouse model (Kdm4b+/-), focusing on neuroanatomical changes. In mutant mice, the total brain volume was significantly reduced with decreased size of the hippocampal dentate gyrus, partial agenesis of the corpus callosum, and ventriculomegaly. This report demonstrates that variants in KDM4B are associated with GDD/ intellectual disability and neuroanatomical defects. Our findings suggest that KDM4B variation leads to a chromatinopathy, broadening the spectrum of this group of Mendelian disorders caused by alterations in epigenetic machinery.


Developmental Disabilities/genetics , Genetic Variation , Jumonji Domain-Containing Histone Demethylases/genetics , Nervous System Malformations/genetics , Animals , Brain/diagnostic imaging , Epigenesis, Genetic , Female , Heterozygote , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Histones/metabolism , Humans , Magnetic Resonance Imaging , Male , Methylation , Mice , Protein Processing, Post-Translational , Seizures/genetics , Signal Transduction
10.
PLoS Genet ; 16(9): e1008916, 2020 09.
Article En | MEDLINE | ID: mdl-32877400

Some imprinted genes exhibit parental origin specific expression bias rather than being transcribed exclusively from one copy. The physiological relevance of this remains poorly understood. In an analysis of brain-specific allele-biased expression, we identified that Trappc9, a cellular trafficking factor, was expressed predominantly (~70%) from the maternally inherited allele. Loss-of-function mutations in human TRAPPC9 cause a rare neurodevelopmental syndrome characterized by microcephaly and obesity. By studying Trappc9 null mice we discovered that homozygous mutant mice showed a reduction in brain size, exploratory activity and social memory, as well as a marked increase in body weight. A role for Trappc9 in energy balance was further supported by increased ad libitum food intake in a child with TRAPPC9 deficiency. Strikingly, heterozygous mice lacking the maternal allele (70% reduced expression) had pathology similar to homozygous mutants, whereas mice lacking the paternal allele (30% reduction) were phenotypically normal. Taken together, we conclude that Trappc9 deficient mice recapitulate key pathological features of TRAPPC9 mutations in humans and identify a role for Trappc9 and its imprinting in controlling brain development and metabolism.


Intercellular Signaling Peptides and Proteins/deficiency , Microcephaly/genetics , Obesity/genetics , Animals , Child , Female , Gene Expression Regulation , Gene Frequency , Genomic Imprinting , Heterozygote , Homozygote , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Maternal Inheritance , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcephaly/metabolism , Mutation , Obesity/metabolism , Phenotype
11.
Nat Commun ; 10(1): 3465, 2019 08 01.
Article En | MEDLINE | ID: mdl-31371714

Brain morphogenesis is an important process contributing to higher-order cognition, however our knowledge about its biological basis is largely incomplete. Here we analyze 118 neuroanatomical parameters in 1,566 mutant mouse lines and identify 198 genes whose disruptions yield NeuroAnatomical Phenotypes (NAPs), mostly affecting structures implicated in brain connectivity. Groups of functionally similar NAP genes participate in pathways involving the cytoskeleton, the cell cycle and the synapse, display distinct fetal and postnatal brain expression dynamics and importantly, their disruption can yield convergent phenotypic patterns. 17% of human unique orthologues of mouse NAP genes are known loci for cognitive dysfunction. The remaining 83% constitute a vast pool of genes newly implicated in brain architecture, providing the largest study of mouse NAP genes and pathways. This offers a complementary resource to human genetic studies and predict that many more genes could be involved in mammalian brain morphogenesis.


Brain , Genetic Association Studies , Morphogenesis/genetics , Neuroanatomy , Neurogenesis/genetics , Animals , Brain/metabolism , Cell Cycle , Cognition , Cytoskeleton , Gene Regulatory Networks , Genes, Lethal/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mutation , Phenotype , Synapses
12.
PLoS Biol ; 17(4): e3000194, 2019 04.
Article En | MEDLINE | ID: mdl-30973865

Adult-onset hearing loss is very common, but we know little about the underlying molecular pathogenesis impeding the development of therapies. We took a genetic approach to identify new molecules involved in hearing loss by screening a large cohort of newly generated mouse mutants using a sensitive electrophysiological test, the auditory brainstem response (ABR). We review here the findings from this screen. Thirty-eight unexpected genes associated with raised thresholds were detected from our unbiased sample of 1,211 genes tested, suggesting extreme genetic heterogeneity. A wide range of auditory pathophysiologies was found, and some mutant lines showed normal development followed by deterioration of responses, revealing new molecular pathways involved in progressive hearing loss. Several of the genes were associated with the range of hearing thresholds in the human population and one, SPNS2, was involved in childhood deafness. The new pathways required for maintenance of hearing discovered by this screen present new therapeutic opportunities.


Auditory Perception/genetics , Evoked Potentials, Auditory, Brain Stem/genetics , Hearing Loss/genetics , Acoustic Stimulation/methods , Adult , Animals , Anion Transport Proteins/genetics , Child , Electrophysiological Phenomena/genetics , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Genetic Association Studies , Hearing/genetics , Hearing Loss/metabolism , Humans , Male , Mice , Mice, Inbred C57BL
13.
Exp Dermatol ; 28(4): 391-394, 2019 04.
Article En | MEDLINE | ID: mdl-29509981

Myosins are molecular motors that are well known for their role in cell movement and contractile functions. Although extensively studied in muscle physiology, little is known about the function of myosins in mammalian skin. As part of the Sanger Institute Mouse Genetics Project, we have identified a role for Myo10 in pigmentation, with a phenotype unlike those of Myo5a or Myo7a. Adult mice homozygous for a disrupted Myo10 allele on a C57BL/6N background displayed a high degree of penetrance for white patches on their abdomen and dorsal surface. Forepaw syndactyly and hind paw syndactyly were also observed in these mice. Tail epidermal wholemounts showed a complete lack of melanocytes in the hair follicles and interfollicular epidermis. Myo10 has previously been implicated in human pigmentation. Our current study reveals involvement of Myo10 in murine skin pigmentation.


Hair Follicle/pathology , Myosins/genetics , Pigmentation Disorders/genetics , Skin Pigmentation/genetics , Alleles , Animals , Female , Gene Expression , Hair Color/genetics , Male , Melanocytes/metabolism , Melanocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Penetrance , Pigmentation Disorders/pathology , Syndactyly/genetics
14.
J Pathol ; 239(3): 374-83, 2016 07.
Article En | MEDLINE | ID: mdl-27126290

The epidermis is the outermost layer of skin that acts as a barrier to protect the body from the external environment and to control water and heat loss. This barrier function is established through the multistage differentiation of keratinocytes and the presence of bioactive sphingolipids such as ceramides, the levels of which are tightly regulated by a balance of ceramide synthase and ceramidase activities. Here we reveal the essential role of alkaline ceramidase 1 (Acer1) in the skin. Acer1-deficient (Acer1(-/-) ) mice showed elevated levels of ceramide in the skin, aberrant hair shaft cuticle formation and cyclic alopecia. We demonstrate that Acer1 is specifically expressed in differentiated interfollicular epidermis, infundibulum and sebaceous glands and consequently Acer1(-/-) mice have significant alterations in infundibulum and sebaceous gland architecture. Acer1(-/-) skin also shows perturbed hair follicle stem cell compartments. These alterations result in Acer1(-/-) mice showing increased transepidermal water loss and a hypermetabolism phenotype with associated reduction of fat content with age. We conclude that Acer1 is indispensable for mammalian skin homeostasis and whole-body energy homeostasis. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Alkaline Ceramidase/metabolism , Alopecia/enzymology , Ceramides/metabolism , Energy Metabolism , Homeostasis , Alkaline Ceramidase/genetics , Alopecia/physiopathology , Animals , Cell Differentiation , Epidermis/abnormalities , Epidermis/enzymology , Female , Hair Follicle/abnormalities , Hair Follicle/enzymology , Humans , Keratinocytes/enzymology , Keratinocytes/physiology , Male , Mice , Mice, Inbred C57BL , Pituitary Gland/abnormalities , Pituitary Gland/enzymology , Sebaceous Glands/abnormalities , Sebaceous Glands/enzymology , Skin/enzymology , Skin Abnormalities , Sphingolipids/metabolism
15.
PLoS Genet ; 10(10): e1004705, 2014 Oct.
Article En | MEDLINE | ID: mdl-25340873

The skin is a highly regenerative organ which plays critical roles in protecting the body and sensing its environment. Consequently, morbidity and mortality associated with skin defects represent a significant health issue. To identify genes important in skin development and homeostasis, we have applied a high throughput, multi-parameter phenotype screen to the conditional targeted mutant mice generated by the Wellcome Trust Sanger Institute's Mouse Genetics Project (Sanger-MGP). A total of 562 different mouse lines were subjected to a variety of tests assessing cutaneous expression, macroscopic clinical disease, histological change, hair follicle cycling, and aberrant marker expression. Cutaneous lesions were associated with mutations in 23 different genes. Many of these were not previously associated with skin disease in the organ (Mysm1, Vangl1, Trpc4ap, Nom1, Sparc, Farp2, and Prkab1), while others were ascribed new cutaneous functions on the basis of the screening approach (Krt76, Lrig1, Myo5a, Nsun2, and Nf1). The integration of these skin specific screening protocols into the Sanger-MGP primary phenotyping pipelines marks the largest reported reverse genetic screen undertaken in any organ and defines approaches to maximise the productivity of future projects of this nature, while flagging genes for further characterisation.


Mutation/genetics , Phenotype , Skin Physiological Phenomena/genetics , Animals , Embryonic Stem Cells , Hair Follicle/metabolism , Hair Follicle/physiology , Mice , Reverse Genetics
16.
Nat Commun ; 5: 3540, 2014 Apr 11.
Article En | MEDLINE | ID: mdl-24721909

Permanent stop-and-shop large-scale mouse mutant resources provide an excellent platform to decipher tissue phenogenomics. Here we analyse skin from 538 knockout mouse mutants generated by the Sanger Institute Mouse Genetics Project. We optimize immunolabelling of tail epidermal wholemounts to allow systematic annotation of hair follicle, sebaceous gland and interfollicular epidermal abnormalities using ontology terms from the Mammalian Phenotype Ontology. Of the 50 mutants with an epidermal phenotype, 9 map to human genetic conditions with skin abnormalities. Some mutant genes are expressed in the skin, whereas others are not, indicating systemic effects. One phenotype is affected by diet and several are incompletely penetrant. In-depth analysis of three mutants, Krt76, Myo5a (a model of human Griscelli syndrome) and Mysm1, provides validation of the screen. Our study is the first large-scale genome-wide tissue phenotype screen from the International Knockout Mouse Consortium and provides an open access resource for the scientific community.


Phenotype , Reverse Genetics/methods , Skin , Animals , Databases, Genetic , Female , Humans , Male , Mice , Mice, Knockout , Mutation
17.
PLoS One ; 8(3): e58156, 2013.
Article En | MEDLINE | ID: mdl-23516444

Otitis media is a common reason for hearing loss, especially in children. Otitis media is a multifactorial disease and environmental factors, anatomic dysmorphology and genetic predisposition can all contribute to its pathogenesis. However, the reasons for the variable susceptibility to otitis media are elusive. MCPH1 mutations cause primary microcephaly in humans. So far, no hearing impairment has been reported either in the MCPH1 patients or mouse models with Mcph1 deficiency. In this study, Mcph1-deficient (Mcph1(tm1a) (/tm1a) ) mice were produced using embryonic stem cells with a targeted mutation by the Sanger Institute's Mouse Genetics Project. Auditory brainstem response measurements revealed that Mcph1(tm1a) (/tm1a) mice had mild to moderate hearing impairment with around 70% penetrance. We found otitis media with effusion in the hearing-impaired Mcph1(tm1a) (/tm1a) mice by anatomic and histological examinations. Expression of Mcph1 in the epithelial cells of middle ear cavities supported its involvement in the development of otitis media. Other defects of Mcph1(tm1a) (/tm1a) mice included small skull sizes, increased micronuclei in red blood cells, increased B cells and ocular abnormalities. These findings not only recapitulated the defects found in other Mcph1-deficient mice or MCPH1 patients, but also revealed an unexpected phenotype, otitis media with hearing impairment, which suggests Mcph1 is a new gene underlying genetic predisposition to otitis media.


Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Mice , Otitis Media/genetics , Animals , Antibodies, Bacterial/immunology , B-Lymphocytes/immunology , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/metabolism , Cytoskeletal Proteins , Ear, Inner/pathology , Ear, Inner/ultrastructure , Ear, Middle/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Gene Expression , Gene Order , Gene Targeting , Genetic Vectors/genetics , Genomic Instability , Genotype , Hearing Loss/diagnosis , Hearing Loss/genetics , Humans , Mice, Knockout , Mutation , Otitis Media/etiology , Otitis Media/pathology , Otitis Media/physiopathology , Skull/pathology
18.
Ann N Y Acad Sci ; 1139: 151-63, 2008 Oct.
Article En | MEDLINE | ID: mdl-18991859

Adult animals treated with high doses of MDMA ("ecstasy") either on a single day or for several consecutive days show numerous behavioral changes as well as persistent reductions in brain serotonin (5-HT) concentrations and 5-HT transporter (SERT) protein expression. However, such dosing regimens do not adequately mimic the intermittent use patterns commonly seen in adolescent recreational ecstasy users. We have developed and characterized a rat model of intermittent adolescent MDMA exposure that simulates many of the features of human weekend use. Animals treated with our dosing regimen experience only small increases in core body temperature, and their plasma MDMA levels compare favorably with the levels reported for heavy ecstasy users under naturalistic conditions when species differences in drug clearance rates are taken into account. Intermittent adolescent MDMA exposure causes later deficits in object-recognition memory, increased impulsivity in the elevated plus-maze, and reduced sensitivity to a 5-HT(1A) agonist challenge. SERT-immunoreactive fiber density is significantly reduced in the hippocampus but not the neocortex, suggesting that the hippocampus may be particularly vulnerable to moderate MDMA exposure during adolescence. Finally, adolescent MDMA-treated animals are protected (i.e., show tolerance) against the neurotoxic and depressant effects of a subsequent MDMA "binge" challenge. We believe that the present animal model has important clinical relevance based on the similarities between the model and the reported effects of regular ecstasy use.


Behavior, Animal/drug effects , Models, Animal , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Agents/pharmacology , 3,4-Methylenedioxyamphetamine/metabolism , 3,4-Methylenedioxyamphetamine/pharmacology , Adolescent , Adult , Animals , Anxiety , Body Temperature , Disruptive, Impulse Control, and Conduct Disorders , Humans , Memory/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Rats , Serotonin/metabolism , Serotonin Agents/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Substance-Related Disorders , Young Adult
...