Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Semin Neurol ; 43(4): 645-660, 2023 08.
Article in English | MEDLINE | ID: mdl-37586397

ABSTRACT

Gastrointestinal symptoms are common in most forms of neurodevelopment disorders (NDDs) such as in autism spectrum disorders (ASD). The current patient-reported outcome measures with validated questionnaires used in the general population of children without NDDS cannot be used in the autistic individuals. We explore here the multifactorial pathophysiology of ASD and the role of genetics and the environment in this disease spectrum and focus instead on possible diagnostics that could provide future objective insight into the connection of the gut-brain-microbiome in this disease entity. We provide our own data from both humans and a zebrafish model of ASD called Phelan-McDermid Syndrome. We hope that this review highlights the gaps in our current knowledge on many of these profound NDDs and that it provides a future framework upon which clinicians and researchers can build and network with other interested multidisciplinary specialties.


Subject(s)
Autism Spectrum Disorder , Chromosome Disorders , Gastrointestinal Diseases , Neurodevelopmental Disorders , Child , Animals , Humans , Zebrafish , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Gastrointestinal Diseases/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics
2.
Am J Hum Genet ; 98(4): 597-614, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27040688

ABSTRACT

Abnormal protein aggregation is observed in an expanding number of neurodegenerative diseases. Here, we describe a mechanism for intracellular toxic protein aggregation induced by an unusual mutation event in families affected by axonal neuropathy. These families carry distinct frameshift variants in NEFH (neurofilament heavy), leading to a loss of the terminating codon and translation of the 3' UTR into an extra 40 amino acids. In silico aggregation prediction suggested the terminal 20 residues of the altered NEFH to be amyloidogenic, which we confirmed experimentally by serial deletion analysis. The presence of this amyloidogenic motif fused to NEFH caused prominent and toxic protein aggregates in transfected cells and disrupted motor neurons in zebrafish. We identified a similar aggregation-inducing mechanism in NEFL (neurofilament light) and FUS (fused in sarcoma), in which mutations are known to cause aggregation in Charcot-Marie-Tooth disease and amyotrophic lateral sclerosis, respectively. In summary, we present a protein-aggregation-triggering mechanism that should be taken into consideration during the evaluation of stop-loss variants.


Subject(s)
3' Untranslated Regions/genetics , Axons/pathology , Intermediate Filaments/genetics , Motor Neurons/pathology , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Line , Charcot-Marie-Tooth Disease/genetics , Frameshift Mutation , Humans , Intermediate Filaments/metabolism , Mice , Molecular Sequence Data , Motor Neurons/metabolism , Mutation , Pedigree , Zebrafish/genetics
3.
Hum Mol Genet ; 24(14): 4006-23, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25882707

ABSTRACT

Despite significant progress in the genetics of autism spectrum disorder (ASD), how genetic mutations translate to the behavioral changes characteristic of ASD remains largely unknown. ASD affects 1-2% of children and adults, and is characterized by deficits in verbal and non-verbal communication, and social interactions, as well as the presence of repetitive behaviors and/or stereotyped interests. ASD is clinically and etiologically heterogeneous, with a strong genetic component. Here, we present functional data from syngap1 and shank3 zebrafish loss-of-function models of ASD. SYNGAP1, a synaptic Ras GTPase activating protein, and SHANK3, a synaptic scaffolding protein, were chosen because of mounting evidence that haploinsufficiency in these genes is highly penetrant for ASD and intellectual disability (ID). Orthologs of both SYNGAP1 and SHANK3 are duplicated in the zebrafish genome and we find that all four transcripts (syngap1a, syngap1b, shank3a and shank3b) are expressed at the earliest stages of nervous system development with pronounced expression in the larval brain. Consistent with early expression of these genes, knockdown of syngap1b or shank3a cause common embryonic phenotypes including delayed mid- and hindbrain development, disruptions in motor behaviors that manifest as unproductive swim attempts, and spontaneous, seizure-like behaviors. Our findings indicate that both syngap1b and shank3a play novel roles in morphogenesis resulting in common brain and behavioral phenotypes.


Subject(s)
Autism Spectrum Disorder/genetics , Brain/embryology , GTPase-Activating Proteins/genetics , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , ras GTPase-Activating Proteins/genetics , Animals , Databases, Genetic , Embryonic Development , GTPase-Activating Proteins/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Haploinsufficiency , Nerve Tissue Proteins/metabolism , Phenotype , Zebrafish/embryology , Zebrafish Proteins/metabolism , ras GTPase-Activating Proteins/metabolism
4.
Am J Hum Genet ; 91(5): 872-82, 2012 11 02.
Article in English | MEDLINE | ID: mdl-23122586

ABSTRACT

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Mutation , Adolescent , Animals , Child, Preschool , Chromosome Aberrations , Cochlea/metabolism , Cochlea/pathology , Exome , Gene Expression Profiling , Gene Knockdown Techniques , Hearing Loss, Sensorineural/diagnosis , Humans , INDEL Mutation , Male , Mice , Polymorphism, Single Nucleotide , Rats , Zebrafish
5.
J Neurosci ; 33(17): 7384-92, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23616544

ABSTRACT

A long-held tenet of neuromuscular transmission is that calcium-dependent neurotransmitter release is mediated by N-type calcium channels in frog but P/Q-type channels in mammals. The N-type assignment in frog is based principally on pharmacological sensitivity to ω-conotoxin GVIA. Our studies show that zebrafish neuromuscular transmission is also sensitive to ω-conotoxin GVIA. However, positional cloning of a mutant line with compromised neuromuscular function identified a mutation in a P/Q- rather than N-type channel. Cloning and heterologous expression of this P/Q-type channel confirmed a block by ω-conotoxin GVIA raising the likelihood that all vertebrates, including frog, use the P/Q-type calcium channel for neuromuscular transmission. In addition, our P/Q defective mutant line offered a means of testing the ability of roscovitine, known to potentiate frog neuromuscular transmission, to mediate behavioral and functional rescue. Acute treatment led to rapid improvement of both, pointing to potential therapeutic benefit for myasthenic disorders involving calcium channel dysfunction.


Subject(s)
Calcium Channels, P-Type/physiology , Calcium Channels, Q-Type/physiology , Neuromuscular Junction/physiology , Synaptic Transmission/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Calcium Channels/physiology , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/physiology , Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Cloning, Molecular , HEK293 Cells , Humans , Molecular Sequence Data , Mutation/physiology , Neuromuscular Junction/genetics , Synaptic Transmission/genetics , Zebrafish
6.
Am J Hum Genet ; 88(2): 201-6, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21295283

ABSTRACT

Increasingly, mutations in genes causing Mendelian disease will be supported by individual and small families only; however, exome sequencing studies have thus far focused on syndromic phenotypes characterized by low locus heterogeneity. In contrast, retinitis pigmentosa (RP) is caused by >50 known genes, which still explain only half of the clinical cases. In a single, one-generation, nonsyndromic RP family, we have identified a gene, dehydrodolichol diphosphate synthase (DHDDS), demonstrating the power of combining whole-exome sequencing with rapid in vivo studies. DHDDS is a highly conserved essential enzyme for dolichol synthesis, permitting global N-linked glycosylation. Zebrafish studies showed virtually identical photoreceptor defects as observed with N-linked glycosylation-interfering mutations in the light-sensing protein rhodopsin. The identified Lys42Glu variant likely arose from an ancestral founder, because eight of the nine identified alleles in 27,174 control chromosomes were of confirmed Ashkenazi Jewish ethnicity. These findings demonstrate the power of exome sequencing linked to functional studies when faced with challenging study designs and, importantly, link RP to the pathways of N-linked glycosylation, which promise new avenues for therapeutic interventions.


Subject(s)
Alkyl and Aryl Transferases/genetics , Exons/genetics , Genetic Variation/genetics , Mutation/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , Animals , Dolichols/analogs & derivatives , Dolichols/metabolism , Female , Genes, Dominant , Glycosylation , Humans , Male , Pedigree , Phenotype , Polymerase Chain Reaction , Sequence Analysis, DNA , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
Adv Exp Med Biol ; 801: 165-70, 2014.
Article in English | MEDLINE | ID: mdl-24664694

ABSTRACT

A single-nucleotide mutation in the gene that encodes DHDDS has been identified by whole exome sequencing as the cause of the non-syndromic recessive retinitis pigmentosa (RP) in a family of Ashkenazi Jewish origin in which three of the four siblings have early onset retinal degeneration. The peripheral retinal degeneration in the affected siblings was evident in the initial examination in 1992 and only one had detectable electroretinogram (ERG) that suggested cone-rod dysfunction. The pigmentary retinal degeneration subsequently progressed rapidly. The identified mutation changes the highly conserved residue Lys42 to Glu, resulting in lower catalytic efficiency. Patterns of plasma transferrin isoelectric focusing gel were normal in all family members, indicating no significant abnormality in protein glycosylation. Dolichols have been shown to influence the fluidity and of the membrane and promote vesicle fusion. Considering that photoreceptor outer segments contain stacks of membrane discs, we believe that the mutation may lead to low dolichol levels in photoreceptor outer segments, resulting in unstable membrane structure that leads to photoreceptor degeneration.


Subject(s)
Alkyl and Aryl Transferases/genetics , Point Mutation , Retina/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Adult , Alkyl and Aryl Transferases/metabolism , Dolichols/metabolism , Female , Genes, Recessive , Humans , Jews/genetics , Male , Pedigree , Retinitis Pigmentosa/metabolism
8.
Adv Exp Med Biol ; 801: 543-50, 2014.
Article in English | MEDLINE | ID: mdl-24664742

ABSTRACT

A mutation in the dehydrodolichol diphosphate synthase (DHDDS) was recently identified as the cause of a subtype of recessive retinitis pigmentosa (RP). Molecular modeling indicates that this mutation could result in low enzymatic efficiency of DHDDS. To investigate the possible link between insufficient DHDDS activity and photoreceptor degeneration, the expression of DHDDS was knocked down by morpholino oligonucleotides (MO) injected into zebrafish one cell embryos. The general appearance and behavior of 4-day-old MO-injected fish were normal, but they failed to respond to light-off, suggesting loss of visual function. Morphological analysis showed that photoreceptor outer segments in retinas of MO-injected fish are very short and in many cases completely missing. Peanut agglutinin (PNA) staining confirmed the absence of cone outer segments. These results demonstrate that suppression of DHDDS expression in zebrafish leads to the loss of photoreceptor outer segments and visual function. These results support the hypothesis that insufficient DHDDS function leads to retinal degeneration.


Subject(s)
Alkyl and Aryl Transferases/genetics , Disease Models, Animal , Retinal Degeneration/genetics , Retinal Photoreceptor Cell Outer Segment/physiology , Zebrafish Proteins/genetics , Zebrafish/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Gene Knockdown Techniques , Introns/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Photoreceptor Cell Outer Segment/metabolism , Retinal Photoreceptor Cell Outer Segment/pathology , Zebrafish Proteins/metabolism
9.
Neurobiol Dis ; 60: 139-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24029548

ABSTRACT

Startle disease is an inherited neurological disorder that causes affected individuals to suffer noise- or touch-induced non-epileptic seizures, excessive muscle stiffness and neonatal apnea episodes. Mutations known to cause startle disease have been identified in glycine receptor subunit (GLRA1 and GLRB) and glycine transporter (SLC6A5) genes, which serve essential functions at glycinergic synapses. Despite the significant successes in identifying startle disease mutations, many idiopathic cases remain unresolved. Exome sequencing in these individuals will identify new candidate genes. To validate these candidate disease genes, zebrafish is an ideal choice due to rapid knockdown strategies, accessible embryonic stages, and stereotyped behaviors. The only existing zebrafish model of startle disease, bandoneon (beo), harbors point mutations in glrbb (one of two zebrafish orthologs of human GLRB) that cause compromised glycinergic transmission and touch-induced bilateral muscle contractions. In order to further develop zebrafish as a model for startle disease, we sought to identify common phenotypic outcomes of knocking down zebrafish orthologs of two known startle disease genes, GLRA1 and GLRB, using splice site-targeted morpholinos. Although both morphants were expected to result in phenotypes similar to the zebrafish beo mutant, our direct comparison demonstrated that while both glra1 and glrbb morphants exhibited embryonic spasticity, only glrbb morphants exhibited bilateral contractions characteristic of beo mutants. Likewise, zebrafish over-expressing a dominant startle disease mutation (GlyR α1(R271Q)) exhibited spasticity but not bilateral contractions. Since GlyR ßb can interact with GlyR α subunits 2-4 in addition to GlyR α1, loss of the GlyR ßb subunit may produce more severe phenotypes by affecting multiple GlyR subtypes. Indeed, immunohistochemistry of glra1 morphants suggests that in zebrafish, alternate GlyR α subunits can compensate for the loss of the GlyR α1 subunit. To address the potential for interplay among GlyR subunits during development, we quantified the expression time-course for genes known to be critical to glycinergic synapse function. We found that GlyR α2, α3 and α4a are expressed in the correct temporal pattern and could compensate for the loss of the GlyR α1 subunit. Based on our findings, future studies that aim to model candidate startle disease genes in zebrafish should include measures of spasticity and synaptic development.


Subject(s)
Disease Models, Animal , Phenotype , Receptors, Glycine/genetics , Stiff-Person Syndrome/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Behavior, Animal/physiology , Morpholinos/genetics , Protein Subunits/genetics , Zebrafish/embryology , Zebrafish/physiology
10.
F1000Res ; 12: 84, 2023.
Article in English | MEDLINE | ID: mdl-37868296

ABSTRACT

Background: Altered sensory processing is a pervasive symptom in individuals with Autism Spectrum Disorders (ASD); people with Phelan McDermid syndrome (PMS), in particular, show reduced responses to sensory stimuli. PMS is caused by deletions of the terminal end of chromosome 22 or point mutations in Shank3. People with PMS can present with an array of symptoms including ASD, epilepsy, gastrointestinal distress, and reduced responses to sensory stimuli. People with PMS are often medicated to manage behaviors like aggression and/or self-harm and/or epilepsy, and it remains unclear how these medications might impact perception/sensory processing. Here we test this using zebrafish mutant shank3ab PMS models that likewise show reduced sensory responses in a visual motor response (VMR) assay, in which increased locomotion is triggered by light to dark transitions. Methods: We screened three medications, risperidone, lithium chloride (LiCl), and carbamazepine (CBZ), prescribed to people with PMS and one drug, 2-methyl-6-(phenylethynyl) pyridine (MPEP) tested in rodent models of PMS, for their effects on a sensory-induced behavior in two zebrafish PMS models with frameshift mutations in either the N- or C- termini. To test how pharmacological treatments affect the VMR, we exposed larvae to selected drugs for 24 hours and then quantified their locomotion during four ten-minute cycles of lights on-to-off stimuli. Results: We found that risperidone normalized the VMR in shank3 models. LiCl and CBZ had no effect on the VMR in any of the three genotypes. MPEP reduced the VMR in wildtype (WT) to levels seen in shank3 models but caused no changes in either shank3 model. Finally, shank3 mutants showed resistance to the seizure-inducing drug pentylenetetrazol (PTZ), at a dosage that results in hyperactive swimming in WT zebrafish. Conclusions: Our work shows that the effects of drugs on sensory processing are varied in ways that can be highly genotype- and drug-dependent.


Subject(s)
Chromosome Disorders , Perception , Zebrafish , Animals , Humans , Chromosomes, Human, Pair 22 , Nerve Tissue Proteins/genetics , Risperidone/pharmacology , Zebrafish/genetics , Chromosome Disorders/drug therapy , Chromosome Disorders/genetics , Disease Models, Animal , Lithium Chloride/pharmacology , Carbamazepine/pharmacology , Perception/drug effects
11.
bioRxiv ; 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37786701

ABSTRACT

Background and Aims: SYNGAP1 disorder is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied. Methods: We used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli. Results: We show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab larvae are hyperactive compared to wild type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, with overall movement increasing with the number of mutant syngap1 alleles. Conclusions: Our data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.

12.
Nanoscale ; 14(47): 17607-17624, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36412202

ABSTRACT

The carbon nitride dot (CND) is an emerging carbon-based nanomaterial. It possesses rich surface functional moieties and a carbon nitride core. Spectroscopic data have demonstrated the analogy between CNDs and cytosine/uracil. Recently, it was found that CNDs could interrupt the normal embryogenesis of zebrafish. Modifying CNDs with various nucleobases, especially cytosine, further decreased embryo viability and increased deformities. Physicochemical property characterization demonstrated that adenine- and cytosine-incorporated CNDs are similar but different from guanine-, thymine- and uracil-incorporated CNDs in many properties, morphology, and structure. To investigate the embryogenesis interruption at the cellular level, bare and different nucleobase-incorporated CNDs were applied to normal and cancerous cell lines. A dose-dependent decline was observed in the viability of normal and cancerous cells incubated with cytosine-incorporated CNDs, which matched results from the zebrafish embryogenesis experiment. In addition, nucleobase-incorporated CNDs were observed to enter cell nuclei, demonstrating a possibility of CND-DNA interactions. CNDs modified by complementary nucleobases could bind each other via hydrogen bonds, which suggests nucleobase-incorporated CNDs can potentially bind the complementary nucleobases in a DNA double helix. Nonetheless, neither bare nor nucleobase-incorporated CNDs were observed to intervene in the amplification of the zebrafish polymerase-alpha 1 gene in quantitative polymerase chain reactions. Thus, in conclusion, the embryogenesis interruption by bare and nucleobase-incorporated CNDs might not be a consequence of CND-DNA interactions during DNA replication. Instead, CND-Ca2+ interactions offer a plausible mechanism that hindered cell proliferation and zebrafish embryogenesis originating from disturbed Ca2+ homeostasis by CNDs. Eventually, the hypothesis that raw or nucleobase-incorporated CNDs can be nucleobase analogs proved to be invalid.


Subject(s)
Cytosine , Zebrafish , Animals , Uracil
13.
Front Cell Dev Biol ; 9: 662916, 2021.
Article in English | MEDLINE | ID: mdl-33937265

ABSTRACT

Research involving autism spectrum disorder (ASD) most frequently focuses on its key diagnostic criteria: restricted interests and repetitive behaviors, altered sensory perception, and communication impairments. These core criteria, however, are often accompanied by numerous comorbidities, many of which result in severe negative impacts on quality of life, including seizures, epilepsy, sleep disturbance, hypotonia, and GI distress. While ASD is a clinically heterogeneous disorder, gastrointestinal (GI) distress is among the most prevalent co-occurring symptom complex, manifesting in upward of 70% of all individuals with ASD. Consistent with this high prevalence, over a dozen family foundations that represent genetically distinct, molecularly defined forms of ASD have identified GI symptoms as an understudied area with significant negative impacts on quality of life for both individuals and their caregivers. Moreover, GI symptoms are also correlated with more pronounced irritability, social withdrawal, stereotypy, hyperactivity, and sleep disturbances, suggesting that they may exacerbate the defining behavioral symptoms of ASD. Despite these facts (and to the detriment of the community), GI distress remains largely unaddressed by ASD research and is frequently regarded as a symptomatic outcome rather than a potential contributory factor to the behavioral symptoms. Allowing for examination of both ASD's impact on the central nervous system (CNS) as well as its impact on the GI tract and the associated microbiome, the zebrafish has recently emerged as a powerful tool to study ASD. This is in no small part due to the advantages zebrafish present as a model system: their precocious development, their small transparent larval form, and their parallels with humans in genetics and physiology. While ASD research centered on the CNS has leveraged these advantages, there has been a critical lack of GI-centric ASD research in zebrafish models, making a holistic view of the gut-brain-microbiome axis incomplete. Similarly, high-throughput ASD drug screens have recently been developed but primarily focus on CNS and behavioral impacts while potential GI impacts have not been investigated. In this review, we aim to explore the great promise of the zebrafish model for elucidating the roles of the gut-brain-microbiome axis in ASD.

14.
Commun Biol ; 4(1): 1411, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921227

ABSTRACT

People with Phelan-McDermid Syndrome, caused by mutations in the SHANK3 gene, commonly exhibit reduced responses to sensory stimuli; yet the changes in brain-wide activity that link these symptoms to mutations in the shank3 gene remain unknown. Here we quantify movement in response to sudden darkness in larvae of two shank3 zebrafish mutant models and show that both models exhibit dampened responses to this stimulus. Using brain-wide activity mapping, we find that shank3-/- light-sensing brain regions show normal levels of activity while sensorimotor integration and motor regions are less active. Specifically restoring Shank3 function in a sensorimotor nucleus of the rostral brainstem enables the shank3-/- model to respond like wild-type. In sum, we find that reduced sensory responsiveness in shank3-/- models is associated with reduced activity in sensory processing brain regions and can be rescued by restoring Shank3 function in the rostral brainstem. These studies highlight the importance of Shank3 function in the rostral brainstem for integrating sensory inputs to generate behavioral adaptations to changing sensory stimuli.


Subject(s)
Autistic Disorder/genetics , Brain Stem/physiology , Chromosome Disorders/genetics , Nerve Tissue Proteins/genetics , Zebrafish Proteins/genetics , Animals , Autistic Disorder/physiopathology , Chromosome Deletion , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 22/metabolism , Disease Models, Animal , Mutation , Nerve Tissue Proteins/metabolism , Zebrafish , Zebrafish Proteins/metabolism
15.
Sci Rep ; 11(1): 3148, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542258

ABSTRACT

Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.


Subject(s)
Delayed Emergence from Anesthesia/genetics , Glycine Plasma Membrane Transport Proteins/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/genetics , Neurons/metabolism , Preoptic Area/metabolism , Zebrafish Proteins/genetics , Aminobenzoates , Anesthesia, General , Anesthetics , Animals , Animals, Genetically Modified , Craniotomy , Delayed Emergence from Anesthesia/metabolism , Delayed Emergence from Anesthesia/physiopathology , Delayed Emergence from Anesthesia/prevention & control , Disease Models, Animal , Gene Expression , Glycine/pharmacology , Glycine Plasma Membrane Transport Proteins/deficiency , Hyperglycinemia, Nonketotic/drug therapy , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/physiopathology , Ketamine , Locomotion/physiology , Neurons/drug effects , Neurons/pathology , Preoptic Area/drug effects , Preoptic Area/pathology , Propofol , Zebrafish , Zebrafish Proteins/deficiency , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
16.
PLoS One ; 15(3): e0230566, 2020.
Article in English | MEDLINE | ID: mdl-32208444

ABSTRACT

A phenomenon of genetic compensation is commonly observed when an organism with a disease-bearing mutation shows incomplete penetrance of the disease phenotype. Such incomplete phenotypic penetrance, or genetic compensation, is more commonly found in stable knockout models, rather than transient knockdown models. As such, these incidents present a challenge for the disease modeling field, although a deeper understanding of genetic compensation may also hold the key for novel therapeutic interventions. In our study we created a knockout model of slc25a46 gene, which is a recently discovered important player in mitochondrial dynamics, and deleterious mutations in which are known to cause peripheral neuropathy, optic atrophy and cerebellar ataxia. We report a case of genetic compensation in a stable slc25a46 homozygous zebrafish mutant (hereafter referred as "mutant"), in contrast to a penetrant disease phenotype in the first generation (F0) slc25a46 mosaic mutant (hereafter referred as "crispant"), generated with CRISPR/Cas-9 technology. We show that the crispant phenotype is specific and rescuable. By performing mRNA sequencing, we define significant changes in slc25a46 mutant's gene expression profile, which are largely absent in crispants. We find that among the most significantly altered mRNAs, anxa6 gene stands out as a functionally relevant player in mitochondrial dynamics. We also find that our genetic compensation case does not arise from mechanisms driven by mutant mRNA decay. Our study contributes to the growing evidence of the genetic compensation phenomenon and presents novel insights about Slc25a46 function. Furthermore, our study provides the evidence for the efficiency of F0 CRISPR screens for disease candidate genes, which may be used to advance the field of functional genetics.


Subject(s)
CRISPR-Cas Systems , Mitochondrial Membrane Transport Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cerebellar Ataxia/genetics , Disease Models, Animal , Female , Gene Targeting , Male , Mutagenesis , Mutation , Optic Atrophy/genetics , Peripheral Nervous System Diseases/genetics
17.
Nat Genet ; 52(5): 473-481, 2020 05.
Article in English | MEDLINE | ID: mdl-32367058

ABSTRACT

Here we report biallelic mutations in the sorbitol dehydrogenase gene (SORD) as the most frequent recessive form of hereditary neuropathy. We identified 45 individuals from 38 families across multiple ancestries carrying the nonsense c.757delG (p.Ala253GlnfsTer27) variant in SORD, in either a homozygous or compound heterozygous state. SORD is an enzyme that converts sorbitol into fructose in the two-step polyol pathway previously implicated in diabetic neuropathy. In patient-derived fibroblasts, we found a complete loss of SORD protein and increased intracellular sorbitol. Furthermore, the serum fasting sorbitol levels in patients were dramatically increased. In Drosophila, loss of SORD orthologs caused synaptic degeneration and progressive motor impairment. Reducing the polyol influx by treatment with aldose reductase inhibitors normalized intracellular sorbitol levels in patient-derived fibroblasts and in Drosophila, and also dramatically ameliorated motor and eye phenotypes. Together, these findings establish a novel and potentially treatable cause of neuropathy and may contribute to a better understanding of the pathophysiology of diabetes.

19.
Mol Autism ; 10: 3, 2019.
Article in English | MEDLINE | ID: mdl-30733854

ABSTRACT

Background and aims: Autism spectrum disorder (ASD) is currently estimated to affect more than 1% of the world population. For people with ASD, gastrointestinal (GI) distress is a commonly reported but a poorly understood co-occurring symptom. Here, we investigate the physiological basis for GI distress in ASD by studying gut function in a zebrafish model of Phelan-McDermid syndrome (PMS), a condition caused by mutations in the SHANK3 gene. Methods: To generate a zebrafish model of PMS, we used CRISPR/Cas9 to introduce clinically related C-terminal frameshift mutations in shank3a and shank3b zebrafish paralogues (shank3abΔC). Because PMS is caused by SHANK3 haploinsufficiency, we assessed the digestive tract (DT) structure and function in zebrafish shank3abΔC+/- heterozygotes. Human SHANK3 mRNA was then used to rescue DT phenotypes in larval zebrafish. Results: Significantly slower rates of DT peristaltic contractions (p < 0.001) with correspondingly prolonged passage time (p < 0.004) occurred in shank3abΔC+/- mutants. Rescue injections of mRNA encoding the longest human SHANK3 isoform into shank3abΔC+/- mutants produced larvae with intestinal bulb emptying similar to wild type (WT), but still deficits in posterior intestinal motility. Serotonin-positive enteroendocrine cells (EECs) were significantly reduced in both shank3abΔC+/- and shank3abΔC-/- mutants (p < 0.05) while enteric neuron counts and overall structure of the DT epithelium, including goblet cell number, were unaffected in shank3abΔC+/- larvae. Conclusions: Our data and rescue experiments support mutations in SHANK3 as causal for GI transit and motility abnormalities. Reductions in serotonin-positive EECs and serotonin-filled ENS boutons suggest an endocrine/neural component to this dysmotility. This is the first study to date demonstrating DT dysmotility in a zebrafish single gene mutant model of ASD.


Subject(s)
Autistic Disorder/genetics , Gastrointestinal Motility , Nerve Tissue Proteins/genetics , Zebrafish Proteins/genetics , Animals , Autistic Disorder/physiopathology , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Enteroendocrine Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/growth & development , Intestines/physiology , Mutation , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Serotonin/metabolism , Zebrafish
20.
Colloids Surf B Biointerfaces ; 176: 488-493, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30690384

ABSTRACT

Drug traversal across the blood-brain barrier has come under increasing scrutiny recently, particularly concerning the treatment of sicknesses, such as brain cancer and Alzheimer's disease. Most therapies and medicines are limited due to their inability to cross this barrier, reducing treatment options for maladies affecting the brain. Carbon dots show promise as drug carriers, but they experience the same limitations regarding crossing the blood-brain barrier as many small molecules do. If carbon dots can be prepared from a precursor that can cross the blood-brain barrier, there is a chance that the remaining original precursor molecule can attach to the carbon dot surface and lead the system into the brain. Herein, tryptophan carbon dots were synthesized with the strategy of using tryptophan as an amino acid for crossing the blood-brain barrier via LAT1 transporter-mediated endocytosis. Two types of carbon dots were synthesized using tryptophan and two different nitrogen dopants: urea and 1,2-ethylenediamine. Carbon dots made using these precursors show excitation wavelength-dependent emission, low toxicity, and have been observed inside the central nervous system of zebrafish (Danio rerio). The proposed mechanism for these carbon dots abilities to cross the blood-brain barrier concerns residual tryptophan molecules which attach to the carbon dots surface, enabling them to be recognized by the LAT1 transporter. The role of carbon dots for transport open promising avenues for drug delivery and imaging in the brain.


Subject(s)
Blood-Brain Barrier/metabolism , Carbon/metabolism , Tryptophan/metabolism , Animals , Animals, Genetically Modified , Ethylenediamines/metabolism , Spectrum Analysis , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL