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










Publication year range
1.
J Autism Dev Disord ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37642871

ABSTRACT

Objective, quantitative measures of caregiver-child interaction during play are needed to complement caregiver or examiner ratings for clinical assessment and tracking intervention responses. In this exploratory study, we examined the feasibility of using automated video tracking, Noldus EthoVision XT, to measure 159 2-to-7-year-old autistic children's patterns of movement during play-based, caregiver-child interactions and examined their associations with standard clinical measures and human observational coding of caregiver-child joint engagement. Results revealed that autistic children who exhibited higher durations and velocity of movement were, on average, younger, had lower cognitive abilities, greater autism-related features, spent less time attending to the caregiver, and showed lower levels of joint engagement. After adjusting for age and nonverbal cognitive abilities, we found that children who remained in close proximity to their caregiver were more likely to engage in joint engagement that required support from the caregiver. These findings suggest that video tracking offers promise as a scalable, quantitative, and relevant measure of autism-related behaviors.

2.
Autism ; 27(8): 2530-2541, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37151032

ABSTRACT

LAY ABSTRACT: Play-based observations allow researchers to observe autistic children across a wide range of ages and skills. We recorded autistic children playing with toys in the center of a room and at a corner table while a caregiver remained seated off to the side and used video tracking technology to track children's movement and location. We examined how time children spent in room regions and whether or not they approached each region during play related to their cognitive, social, communication, and adaptive skills to determine if tracking child movement and location can meaningfully demonstrate clinical variation among autistic children representing a range of ages and skills. One significant finding was that autistic children who spent more time in the toy-containing center of the room had higher cognitive and language abilities, whereas those who spent less time in the center had higher levels of autism-related behaviors. In contrast, children who spent more time in the caregiver region had lower daily living skills and those who were quicker to approach the caregiver had lower adaptive behavior and language skills. These findings support the use of movement tracking as a complementary method of measuring clinical differences among autistic children. Furthermore, over 90% of autistic children representing a range of ages and skills in this study provided analyzable play observation data, demonstrating that this method allows autistic children of all levels of support needs to participate in research and demonstrate their social, communication, and attention skills without wearing any devices.

3.
Neuron ; 110(10): 1728-1741.e7, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35294900

ABSTRACT

The architecture whereby activity across many brain regions integrates to encode individual appetitive social behavior remains unknown. Here we measure electrical activity from eight brain regions as mice engage in a social preference assay. We then use machine learning to discover a network that encodes the extent to which individual mice engage another mouse. This network is organized by theta oscillations leading from prelimbic cortex and amygdala that converge on the ventral tegmental area. Network activity is synchronized with cellular firing, and frequency-specific activation of a circuit within this network increases social behavior. Finally, the network generalizes, on a mouse-by-mouse basis, to encode individual differences in social behavior in healthy animals but fails to encode individual behavior in a 'high confidence' genetic model of autism. Thus, our findings reveal the architecture whereby the brain integrates distributed activity across timescales to encode an appetitive brain state underlying individual differences in social behavior.


Subject(s)
Appetitive Behavior , Brain , Amygdala , Animals , Brain/physiology , Mice , Social Behavior , Ventral Tegmental Area
4.
Pediatrics ; 145(2)2020 02.
Article in English | MEDLINE | ID: mdl-32015180

ABSTRACT

Phenotypic and biological characterization of rare monogenic disorders represents 1 of the most important avenues toward understanding the mechanisms of human disease. Among patients with SH3 and multiple ankyrin repeat domains 3 (SHANK3) mutations, a subset will manifest neurologic regression, psychosis, and mood disorders. However, which patients will be affected, when, and why are important unresolved questions. Authors of recent studies suggest neuronal SHANK3 expression is modulated by both inflammatory and hormonal stimuli. In this case series, we describe 4 independent clinical observations of an immunotherapy responsive phenotype of peripubertal-onset neuropsychiatric regression in 4 girls with pathogenic SHANK3 mutations. Each child exhibited a history of stable, mild-to-moderate lifelong developmental disability until 12 to 14 years of age, at which time each manifested a similar, subacute-onset neurobehavioral syndrome. Symptoms included mutism, hallucinations, insomnia, inconsolable crying, obsessive-compulsive behaviors, loss of self-care, and urinary retention and/or incontinence. Symptoms were relatively refractory to antipsychotic medication but improved after immunomodulatory treatment. All 4 patients exhibited chronic relapsing courses during a period of treatment and follow-up ranging from 3 to 6 years. Two of the 4 girls recovered their premorbid level of functioning. We briefly review the scientific literature to offer a conceptual and molecular framework for understanding these clinical observations. Future clinical and translational investigations in this realm may offer insights into mechanisms and therapies bridging immune function and human behavior.


Subject(s)
Autism Spectrum Disorder/genetics , Developmental Disabilities/genetics , Frameshift Mutation , Immunotherapy/methods , Nerve Tissue Proteins/genetics , Stereotyped Behavior , Adolescent , Aggression/drug effects , Antipsychotic Agents/therapeutic use , Anxiety , Catatonia/drug therapy , Child , Compulsive Behavior/drug therapy , Crying , Female , Hallucinations/drug therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunosuppressive Agents/therapeutic use , Irritable Mood/drug effects , Methylprednisolone/therapeutic use , Mutism/drug therapy , Neuroprotective Agents/therapeutic use , Obsessive-Compulsive Disorder/drug therapy , Recurrence , Self Care , Sleep Initiation and Maintenance Disorders/drug therapy , Stereotyped Behavior/drug effects , Syndrome , Urinary Incontinence , Urinary Retention
5.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518695

ABSTRACT

OXTR modulates a variety of behaviors in mammals, including social memory and recognition. Genetic and epigenetic dysregulation of OXTR has been suggested to be implicated in neuropsychiatric disorders, including autism spectrum disorder (ASD). While the involvement of DNA methylation is suggested, the mechanism underlying epigenetic regulation of OXTR is largely unknown. This has hampered the experimental design and interpretation of the results of epigenetic studies of OXTR in neuropsychiatric disorders. From the generation and characterization of a new line of Tet1 mutant mice - by deleting the largest coding exon 4 (Tet1Δe4) - we discovered for the first time to our knowledge that Oxtr has an array of mRNA isoforms and a complex transcriptional regulation. Select isoforms of Oxtr are significantly reduced in the brain of Tet1Δe4-/- mice. Accordingly, CpG islands of Oxtr are hypermethylated during early development and persist into adulthood. Consistent with the reduced express of OXTR, Tet1Δe4-/- mice display impaired maternal care, social behavior, and synaptic responses to oxytocin stimulation. Our findings elucidate a mechanism mediated by TET1 protein in regulating Oxtr expression by preventing DNA hypermethylation of Oxtr. The discovery of epigenetic dysregulation of Oxtr in TET1-deficient mouse brain supports the necessity of a reassessment of existing findings and a value of future studies of OXTR in neuropsychiatric disorders.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/genetics , Receptors, Oxytocin/genetics , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Behavior, Animal/physiology , Brain/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Exons , Female , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Proto-Oncogene Proteins/metabolism , RNA Isoforms/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Transcriptome
6.
Brain Behav ; 8(11): e01107, 2018 11.
Article in English | MEDLINE | ID: mdl-30317697

ABSTRACT

INTRODUCTION: Several studies have supported the use of enriched environments to prevent the manifestation of ASD-like phenotypes in laboratory rodents. While the translational value of such experiments is unknown, the findings have been relatively consistent across many different models. METHODS: In the current study, we tested the effects of early environmental enrichment on a mouse model of ASD with high construct validity, the Shank3 ∆e4-22 mice our laboratory previously generated and characterized. RESULTS: Contrary to previous reports, we found no benefits of enriched rearing, including no change in repetitive self-grooming or hole-board exploration. Instead, we found that early environmental enrichment increased anxiety-like behavior in all mice regardless of genotype and decreased motor performance specifically in wild-type mice. CONCLUSIONS: Although using a different enrichment protocol may have rescued the phenotypes in our mouse model, these results suggest that a "one-size fits all" approach may not be the best when it comes to behavioral intervention for ASD and underscores the need for effective pharmaceutical development in certain genetic syndromes with severe symptom presentation.


Subject(s)
Autism Spectrum Disorder/psychology , Environment , Animal Husbandry/methods , Animals , Anxiety/etiology , Disease Models, Animal , Exploratory Behavior/physiology , Female , Genotype , Grooming/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/deficiency , Phenotype
7.
Transl Psychiatry ; 8(1): 94, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29700290

ABSTRACT

We previously reported a new line of Shank3 mutant mice which led to a complete loss of Shank3 by deleting exons 4-22 (Δe4-22) globally. Δe4-22 mice display robust ASD-like behaviors including impaired social interaction and communication, increased stereotypical behavior and excessive grooming, and a profound deficit in instrumental learning. However, the anatomical and neural circuitry underlying these behaviors are unknown. We generated mice with Shank3 selectively deleted in forebrain, striatum, and striatal D1 and D2 cells. These mice were used to interrogate the circuit/brain-region and cell-type specific role of Shank3 in the expression of autism-related behaviors. Whole-cell patch recording and biochemical analyses were used to study the synaptic function and molecular changes in specific brain regions. We found perseverative exploratory behaviors in mice with deletion of Shank3 in striatal inhibitory neurons. Conversely, self-grooming induced lesions were observed in mice with deletion of Shank3 in excitatory neurons of forebrain. However, social, communicative, and instrumental learning behaviors were largely unaffected in these mice, unlike what is seen in global Δe4-22 mice. We discovered unique patterns of change for the biochemical and electrophysiological findings in respective brain regions that reflect the complex nature of transcriptional regulation of Shank3. Reductions in Homer1b/c and membrane hyper-excitability were observed in striatal loss of Shank3. By comparison, Shank3 deletion in hippocampal neurons resulted in increased NMDAR-currents and GluN2B-containing NMDARs. These results together suggest that Shank3 may differentially regulate neural circuits that control behavior. Our study supports a dissociation of Shank3 functions in cortical and striatal neurons in ASD-related behaviors, and it illustrates the complexity of neural circuit mechanisms underlying these behaviors.


Subject(s)
Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/psychology , Corpus Striatum/physiopathology , Nerve Tissue Proteins/physiology , Prosencephalon/physiopathology , Animals , Behavior, Animal , Corpus Striatum/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , Hippocampus/physiopathology , Homer Scaffolding Proteins/metabolism , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/genetics , Neurons/physiology , Phenotype , Prosencephalon/metabolism , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Social Behavior , Synapses/metabolism
8.
Neurobiol Dis ; 110: 12-19, 2018 02.
Article in English | MEDLINE | ID: mdl-29097328

ABSTRACT

Epilepsy is prevalent and often medically intractable in Angelman syndrome (AS). AS mouse model (Ube3am-/p+) shows reduced excitatory neurotransmission but lower seizure threshold. The neural mechanism linking the synaptic dysfunction to the seizure remains elusive. We show that the local circuits of Ube3am-/p+in vitro are hyperexcitable and display a unique epileptiform activity, a phenomenon that is reminiscent of the finding in fragile X syndrome (FXS) mouse model. Similar to the FXS model, lovastatin suppressed the epileptiform activity and audiogenic seizures in Ube3am-/p+. The in vitro model of Ube3am-/p+ is valuable for dissection of neural mechanism and epilepsy drug screening in vivo.


Subject(s)
Angelman Syndrome/physiopathology , Anticonvulsants/pharmacology , Disease Models, Animal , Hippocampus/physiopathology , Lovastatin/pharmacology , Angelman Syndrome/complications , Angelman Syndrome/genetics , Animals , Epilepsy/etiology , Epilepsy/physiopathology , Female , Hippocampus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques/methods , Seizures/etiology , Seizures/physiopathology , Ubiquitin-Protein Ligases/genetics
9.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046483

ABSTRACT

Genetic defects in the synaptic scaffolding protein gene, SHANK2, are linked to a variety of neuropsychiatric disorders, including autism spectrum disorders, schizophrenia, intellectual disability, and bipolar disorder, but the molecular mechanisms underlying the pleotropic effects of SHANK2 mutations are poorly understood. We generated and characterized a line of Shank2 mutant mice by deleting exon 24 (Δe24). Shank2Δe24-/- mice engage in significantly increased locomotor activity, display abnormal reward-seeking behavior, are anhedonic, have perturbations in circadian rhythms, and show deficits in social and cognitive behaviors. While these phenotypes recapitulate the pleotropic behaviors associated with human SHANK2-related disorders, major behavioral features in these mice are reminiscent of bipolar disorder. For instance, their hyperactivity was augmented with amphetamine but was normalized with the mood stabilizers lithium and valproate. Shank2 deficiency limited to the forebrain recapitulated the bipolar mania phenotype. The composition and functions of NMDA and AMPA receptors were altered at Shank2-deficient synapses, hinting toward the mechanism underlying these behavioral abnormalities. Human genetic findings support construct validity, and the behavioral features in Shank2 Δe24 mice support face and predictive validities of this model for bipolar mania. Further genetic studies to understand the contribution of SHANK2 deficiencies in bipolar disorder are warranted.


Subject(s)
Bipolar Disorder/genetics , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Amphetamine/pharmacology , Anhedonia , Animals , Antimanic Agents/therapeutic use , Behavior, Animal , Central Nervous System Stimulants/pharmacology , Chronobiology Disorders/drug therapy , Chronobiology Disorders/genetics , Cognitive Dysfunction/genetics , Female , Hippocampus/metabolism , Hippocampus/ultrastructure , Lithium Compounds/therapeutic use , Male , Mice , Mice, Knockout , Motor Activity/drug effects , N-Methylaspartate/metabolism , Phenotype , Prosencephalon/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Behavior Disorders/genetics , Synapses/metabolism
10.
Neuron ; 92(6): 1279-1293, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-27916453

ABSTRACT

Abnormal pain sensitivity is commonly associated with autism spectrum disorders (ASDs) and affects the life quality of ASD individuals. SHANK3 deficiency was implicated in ASD and pain dysregulation. Here, we report functional expression of SHANK3 in mouse dorsal root ganglion (DRG) sensory neurons and spinal cord presynaptic terminals. Homozygous and heterozygous Shank3 complete knockout (Δe4-22) results in impaired heat hyperalgesia in inflammatory and neuropathic pain. Specific deletion of Shank3 in Nav1.8-expressing sensory neurons also impairs heat hyperalgesia in homozygous and heterozygous mice. SHANK3 interacts with transient receptor potential subtype V1 (TRPV1) via Proline-rich region and regulates TRPV1 surface expression. Furthermore, capsaicin-induced spontaneous pain, inward currents in DRG neurons, and synaptic currents in spinal cord neurons are all reduced after Shank3 haploinsufficiency. Finally, partial knockdown of SHANK3 expression in human DRG neurons abrogates TRPV1 function. Our findings reveal a peripheral mechanism of SHANK3, which may underlie pain deficits in SHANK3-related ASDs.


Subject(s)
Hyperalgesia/genetics , Nerve Tissue Proteins/genetics , Pain/genetics , Presynaptic Terminals/metabolism , Sensory Receptor Cells/metabolism , TRPV Cation Channels/metabolism , Animals , Behavior, Animal , Blotting, Western , Capsaicin/toxicity , Ganglia, Spinal/cytology , Humans , Hyperalgesia/metabolism , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Knockout , Microfilament Proteins , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nerve Tissue Proteins/metabolism , Neuralgia/genetics , Neuralgia/metabolism , Pain/chemically induced , Pain/metabolism , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Sensory System Agents/toxicity , Spinal Cord/cytology
11.
Nat Commun ; 7: 11459, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27161151

ABSTRACT

Human neuroimaging studies suggest that aberrant neural connectivity underlies behavioural deficits in autism spectrum disorders (ASDs), but the molecular and neural circuit mechanisms underlying ASDs remain elusive. Here, we describe a complete knockout mouse model of the autism-associated Shank3 gene, with a deletion of exons 4-22 (Δe4-22). Both mGluR5-Homer scaffolds and mGluR5-mediated signalling are selectively altered in striatal neurons. These changes are associated with perturbed function at striatal synapses, abnormal brain morphology, aberrant structural connectivity and ASD-like behaviour. In vivo recording reveals that the cortico-striatal-thalamic circuit is tonically hyperactive in mutants, but becomes hypoactive during social behaviour. Manipulation of mGluR5 activity attenuates excessive grooming and instrumental learning differentially, and rescues impaired striatal synaptic plasticity in Δe4-22(-/-) mice. These findings show that deficiency of Shank3 can impair mGluR5-Homer scaffolding, resulting in cortico-striatal circuit abnormalities that underlie deficits in learning and ASD-like behaviours. These data suggest causal links between genetic, molecular, and circuit mechanisms underlying the pathophysiology of ASDs.


Subject(s)
Autism Spectrum Disorder/physiopathology , Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Homer Scaffolding Proteins/metabolism , Nerve Tissue Proteins/deficiency , Receptor, Metabotropic Glutamate 5/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Behavior, Animal , Cerebral Cortex/pathology , Corpus Striatum/pathology , Female , Humans , Long-Term Synaptic Depression , Male , Mice , Mice, Knockout , Microfilament Proteins , Models, Neurological , Nerve Net/pathology , Nerve Net/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Sequence Deletion , Social Behavior
12.
Curr Protoc Pharmacol ; 66: 5.66.1-5.66.26, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25181011

ABSTRACT

This overview describes many well characterized mouse models of autism spectrum disorders (ASDs). Mouse models considered here were selected because they are examples of genetically engineered models where human genetic evidence supports a causative relationship between the targeted mutation and the behavioral phenotype. As the ASD diagnosis is based primarily on behavioral evaluations in humans in the domains of social interaction, communication, and restricted interests, the murine phenotypes analogous to human autistic behaviors are highlighted for the different models and behaviors. Although genetically engineered mouse models with good construct and face validity are valuable for identifying and defining underlying pathophysiological mechanisms and for developing potential therapeutic interventions for the human condition, the translational value of various rodent behavioral assays remains a subject of debate. Significant challenges associated with modeling ASDs in rodents because of the clinical and molecular heterogeneity that characterize this disorder are also considered.


Subject(s)
Autism Spectrum Disorder , Mice, Transgenic , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/psychology , Autistic Disorder/genetics , Behavior, Animal , Disease Models, Animal , Fragile X Syndrome/genetics , Hamartoma Syndrome, Multiple/genetics , Long QT Syndrome/genetics , Mice , Mice, Mutant Strains/genetics , Mice, Transgenic/genetics , Rett Syndrome/genetics , Syndactyly/genetics , Tuberous Sclerosis/genetics
13.
Mol Autism ; 5: 30, 2014.
Article in English | MEDLINE | ID: mdl-25071925

ABSTRACT

BACKGROUND: Considerable clinical heterogeneity has been well documented amongst individuals with autism spectrum disorders (ASD). However, little is known about the biological mechanisms underlying phenotypic diversity. Genetic studies have established a strong causal relationship between ASD and molecular defects in the SHANK3 gene. Individuals with various defects of SHANK3 display considerable clinical heterogeneity. Different lines of Shank3 mutant mice with deletions of different portions of coding exons have been reported recently. Variable synaptic and behavioral phenotypes have been reported in these mice, which makes the interpretations for these data complicated without the full knowledge of the complexity of the Shank3 transcript structure. METHODS: We systematically examined alternative splicing and isoform-specific expression of Shank3 across different brain regions and developmental stages by regular RT-PCR, quantitative real time RT-PCR (q-PCR), and western blot. With these techniques, we also investigated the effects of neuronal activity and epigenetic modulation on alternative splicing and isoform-specific expression of Shank3. We explored the localization and influence on dendritic spine development of different Shank3 isoforms in cultured hippocampal neurons by cellular imaging. RESULTS: The Shank3 gene displayed an extensive array of mRNA and protein isoforms resulting from the combination of multiple intragenic promoters and extensive alternative splicing of coding exons in the mouse brain. The isoform-specific expression and alternative splicing of Shank3 were brain-region/cell-type specific, developmentally regulated, activity-dependent, and involved epigenetic regulation. Different subcellular distribution and differential effects on dendritic spine morphology were observed for different Shank3 isoforms. CONCLUSIONS: Our results indicate a complex transcriptional regulation of Shank3 in mouse brains. Our analysis of select Shank3 isoforms in cultured neurons suggests that different Shank3 isoforms have distinct functions. Therefore, the different types of SHANK3 mutations found in patients with ASD and different exonic deletions of Shank3 in mutant mice are predicted to disrupt selective isoforms and result in distinct dysfunctions at the synapse with possible differential effects on behavior. Our comprehensive data on Shank3 transcriptional regulation thus provides an essential molecular framework to understand the phenotypic diversity in SHANK3 causing ASD and Shank3 mutant mice.

14.
Dev Neurobiol ; 74(2): 123-35, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23536326

ABSTRACT

Despite recent advances in understanding the molecular mechanisms of autism spectrum disorders (ASD), the current treatments for these disorders are mostly focused on behavioral and educational approaches. The considerable clinical and molecular heterogeneity of ASD present a significant challenge to the development of an effective treatment targeting underlying molecular defects. Deficiency of SHANK family genes causing ASD represent an exciting opportunity for developing molecular therapies because of strong genetic evidence for SHANK as causative genes in ASD and the availability of a panel of Shank mutant mouse models. In this article, we review the literature suggesting the potential for developing therapies based on molecular characteristics and discuss several exciting themes that are emerging from studying Shank mutant mice at the molecular level and in terms of synaptic function.


Subject(s)
Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/therapy , Nerve Tissue Proteins/genetics , Animals , Child Development Disorders, Pervasive/physiopathology , Humans , Mutation , Nerve Tissue Proteins/metabolism , Synaptic Transmission , Transcranial Magnetic Stimulation
15.
Korean J Physiol Pharmacol ; 16(6): 369-78, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23269898

ABSTRACT

Analysis of synaptic plasticity together with behavioral and molecular studies have become a popular approach to model autism spectrum disorders in order to gain insight into the pathosphysiological mechanisms and to find therapeutic targets. Abnormalities of specific types of synaptic plasticity have been revealed in numerous genetically modified mice that have molecular construct validity to human autism spectrum disorders. Constrained by the feasibility of technique, the common regions analyzed in most studies are hippocampus and visual cortex. The relevance of the synaptic defects in these regions to the behavioral abnormalities of autistic like behaviors is still a subject of debate. Because the exact regions or circuits responsible for the core features of autistic behaviors in humans are still poorly understood, investigation using region-specific conditional mutant mice may help to provide the insight into the neuroanatomical basis of autism in the future.

16.
Nat Genet ; 42(8): 676-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20657595

ABSTRACT

Deficiency in the serine protease inhibitor LEKTI is the etiological origin of Netherton syndrome, which causes detachment of the stratum corneum and chronic inflammation. Here we show that the membrane protease matriptase initiates Netherton syndrome in a LEKTI-deficient mouse model by premature activation of a pro-kallikrein cascade. Auto-activation of pro-inflammatory pro-kallikrein-related peptidases that are associated with stratum corneum detachment was either low or undetectable, but they were efficiently activated by matriptase. Ablation of matriptase from LEKTI-deficient mice dampened inflammation, eliminated aberrant protease activity, prevented detachment of the stratum corneum, and improved the barrier function of the epidermis. These results uncover a pathogenic matriptase-pro-kallikrein pathway that could operate in several human skin and inflammatory diseases.


Subject(s)
Kallikreins/metabolism , Serine Endopeptidases/metabolism , Animals , Epidermis/metabolism , Epidermis/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Netherton Syndrome , Peptide Hydrolases/metabolism , Serine Proteinase Inhibitors/metabolism , Skin/metabolism , Skin/pathology
17.
Blood ; 116(9): 1593-603, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20466854

ABSTRACT

The urokinase plasminogen activator receptor (uPAR) has emerged as a potential regulator of cell adhesion, cell migration, proliferation, differentiation, and cell survival in multiple physiologic and pathologic contexts. The urokinase plasminogen activator (uPA) was the first identified ligand for uPAR, but elucidation of the specific functions of the uPA-uPAR interaction in vivo has been difficult because uPA has important physiologic functions that are independent of binding to uPAR and because uPAR engages multiple ligands. Here, we developed a new mouse strain (Plau(GFDhu/GFDhu)) in which the interaction between endogenous uPA and uPAR is selectively abrogated, whereas other functions of both the protease and its receptor are retained. Specifically, we introduced 4 amino acid substitutions into the growth factor domain (GFD) of uPA that abrogate uPAR binding while preserving the overall structure of the domain. Analysis of Plau(GFDhu/GFDhu) mice revealed an unanticipated role of the uPA-uPAR interaction in suppressing inflammation secondary to fibrin deposition. In contrast, leukocyte recruitment and tissue regeneration were unaffected by the loss of uPA binding to uPAR. This study identifies a principal in vivo role of the uPA-uPAR interaction in cell-associated fibrinolysis critical for suppression of fibrin accumulation and fibrin-associated inflammation and provides a valuable model for further exploration of this multifunctional receptor.


Subject(s)
Disease Models, Animal , Fibrin/metabolism , Inflammation/prevention & control , Pneumonia/prevention & control , Receptors, Urokinase Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Liver/immunology , Liver/injuries , Liver/metabolism , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/prevention & control , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , RNA, Messenger/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Reverse Transcriptase Polymerase Chain Reaction , Skin Diseases/immunology , Skin Diseases/metabolism , Skin Diseases/prevention & control , Survival Rate , Urokinase-Type Plasminogen Activator/genetics , Wound Healing
18.
Am J Pathol ; 175(4): 1453-63, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19717635

ABSTRACT

A pericellular proteolytic pathway initiated by the transmembrane serine protease matriptase plays a critical role in the terminal differentiation of epidermal tissues. Matriptase is constitutively expressed in multiple other epithelia, suggesting a putative role of this membrane serine protease in general epithelial homeostasis. Here we generated mice with conditional deletion of the St14 gene, encoding matriptase, and show that matriptase indeed is essential for the maintenance of multiple types of epithelia in the mouse. Thus, embryonic or postnatal ablation of St14 in epithelial tissues of diverse origin and function caused severe organ dysfunction, which was often associated with increased permeability, loss of tight junction function, mislocation of tight junction-associated proteins, and generalized epithelial demise. The study reveals that the homeostasis of multiple simple and stratified epithelia is matriptase-dependent, and provides an important animal model for the exploration of this membrane serine protease in a range of physiological and pathological processes.


Subject(s)
Epithelium/enzymology , Protein Processing, Post-Translational , Serine Endopeptidases/metabolism , Aging/drug effects , Aging/pathology , Alleles , Animals , Embryo, Mammalian/drug effects , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Epithelium/drug effects , Gene Deletion , Homeostasis/drug effects , Intestines/drug effects , Intestines/enzymology , Intestines/pathology , Megacolon/enzymology , Megacolon/pathology , Membrane Proteins/metabolism , Mice , Organ Specificity/drug effects , Permeability/drug effects , Protein Processing, Post-Translational/drug effects , Salivary Glands/drug effects , Salivary Glands/enzymology , Salivary Glands/pathology , Serine Endopeptidases/deficiency , Tamoxifen/pharmacology , Tight Junctions/drug effects , Tight Junctions/metabolism
19.
PLoS Genet ; 3(8): e132, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17696612

ABSTRACT

MUS81 is conserved among plants, animals, and fungi and is known to be involved in mitotic DNA damage repair and meiotic recombination. Here we present a functional characterization of the Arabidopsis thaliana homolog AtMUS81, which has a role in both mitotic and meiotic cells. The AtMUS81 transcript is produced in all tissues, but is elevated greater than 9-fold in the anthers and its levels are increased in response to gamma radiation and methyl methanesulfonate treatment. An Atmus81 transfer-DNA insertion mutant shows increased sensitivity to a wide range of DNA-damaging agents, confirming its role in mitotically proliferating cells. To examine its role in meiosis, we employed a pollen tetrad-based visual assay. Data from genetic intervals on Chromosomes 1 and 3 show that Atmus81 mutants have a moderate decrease in meiotic recombination. Importantly, measurements of recombination in a pair of adjacent intervals on Chromosome 5 demonstrate that the remaining crossovers in Atmus81 are interference sensitive, and that interference levels in the Atmus81 mutant are significantly greater than those in wild type. These data are consistent with the hypothesis that AtMUS81 is involved in a secondary subset of meiotic crossovers that are interference insensitive.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/genetics , Crossing Over, Genetic/physiology , Endonucleases/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/isolation & purification , Carcinogens/pharmacology , Cell Survival , DNA Damage/physiology , Endonucleases/genetics , Endonucleases/isolation & purification , Gene Expression Regulation, Plant/drug effects , Meiosis/genetics , Mutagenesis, Insertional , Mutant Proteins/physiology , Plants, Genetically Modified , Pollen/cytology , Pollen/genetics , Recombination, Genetic
20.
Proc Natl Acad Sci U S A ; 104(10): 3913-8, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17360452

ABSTRACT

Recombination, in the form of cross-overs (COs) and gene conversion (GC), is a highly conserved feature of meiosis from fungi to mammals. Recombination helps ensure chromosome segregation and promotes allelic diversity. Lesions in the recombination machinery are often catastrophic for meiosis, resulting in sterility. We have developed a visual assay capable of detecting Cos and GCs and measuring CO interference in Arabidopsis thaliana. This flexible assay utilizes transgene constructs encoding pollen-expressed fluorescent proteins of three different colors in the qrt1 mutant background. By observing the segregation of the fluorescent alleles in 92,489 pollen tetrads, we demonstrate (i) a correlation between developmental position and CO frequency, (ii) a temperature dependence for CO frequency, (iii) the ability to detect meiotic GC events, and (iv) the ability to rapidly assess CO interference.


Subject(s)
Arabidopsis/genetics , Meiosis , Pollen/genetics , Pollen/metabolism , Recombination, Genetic , Alleles , Arabidopsis Proteins/genetics , Fluorescent Dyes/pharmacology , Gene Conversion , Genes, Plant , Genetic Linkage , Heterozygote , Models, Genetic , Mutagenesis , Temperature , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...