Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Birth Defects Res ; 116(1): e2287, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38116905

ABSTRACT

BACKGROUND: Congenital hydrocephalus (CH) is a life-threatening neurological condition that results from an imbalance in production, flow, or absorption of cerebrospinal fluid. Predicted outcomes from in utero diagnosis are frequently unclear. Moreover, conventional treatments consisting primarily of antenatal and postnatal surgeries are often unsuccessful, leading to high mortality rates. Causes of CH can range from secondary insults to germline pathogenic variants, complicating diagnostic processes and treatment outcomes. Currently, an updated summary of CH genetic etiologies in conjunction with clinical testing methodologies is lacking. This review addresses this need by generating a centralized survey of known genetic causes and available molecular tests for CH. METHODS: The scoping review protocol was registered with the Open Science Framework and followed the Arksey and O'Malley framework and the Joanna Briggs Institute methodology. The Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) was utilized to define search guidelines and screening criteria. RESULTS: Our survey revealed a high number of genetic etiologies associated with CH, ranging from single gene variants to multifactorial birth defects, and additionally uncovered diagnostic challenges that are further complicated by changes in testing approaches over the years. Furthermore, we discovered that most of the existing literature consists of case reports, underscoring the need for studies that utilize CH patient research cohorts as well as more mechanistic studies. CONCLUSIONS: The pursuit of such studies will facilitate novel gene discovery while recognizing phenotypic complexity. Addressing these research gaps could ultimately inform evidence-based diagnostic guidelines to improve patient care.


Subject(s)
Hydrocephalus , Female , Pregnancy , Humans , Hydrocephalus/diagnosis , Hydrocephalus/genetics , Germ-Line Mutation , Prenatal Diagnosis
2.
Genetics ; 220(2)2022 02 04.
Article in English | MEDLINE | ID: mdl-34849872

ABSTRACT

L1CAMs are immunoglobulin cell adhesion molecules that function in nervous system development and function. Besides being associated with autism and schizophrenia spectrum disorders, impaired L1CAM function also underlies the X-linked L1 syndrome, which encompasses a group of neurological conditions, including spastic paraplegia and congenital hydrocephalus. Studies on vertebrate and invertebrate L1CAMs established conserved roles that include axon guidance, dendrite morphogenesis, synapse development, and maintenance of neural architecture. We previously identified a genetic interaction between the Caenorhabditis elegans L1CAM encoded by the sax-7 gene and RAB-3, a GTPase that functions in synaptic neurotransmission; rab-3; sax-7 mutant animals exhibit synthetic locomotion abnormalities and neuronal dysfunction. Here, we show that this synergism also occurs when loss of SAX-7 is combined with mutants of other genes encoding key players of the synaptic vesicle (SV) cycle. In contrast, sax-7 does not interact with genes that function in synaptogenesis. These findings suggest a postdevelopmental role for sax-7 in the regulation of synaptic activity. To assess this possibility, we conducted electrophysiological recordings and ultrastructural analyses at neuromuscular junctions; these analyses did not reveal obvious synaptic abnormalities. Lastly, based on a forward genetic screen for suppressors of the rab-3; sax-7 synthetic phenotypes, we determined that mutants in the ERK Mitogen-activated Protein Kinase (MAPK) pathway can suppress the rab-3; sax-7 locomotion defects. Moreover, we established that Erk signaling acts in a subset of cholinergic neurons in the head to promote coordinated locomotion. In combination, these results suggest a modulatory role for Erk MAPK in L1CAM-dependent locomotion in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins , Neural Cell Adhesion Molecule L1 , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cholinergic Neurons/metabolism , Locomotion , Mitogen-Activated Protein Kinases/genetics , Mutation , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecules/genetics
3.
Development ; 143(7): 1182-91, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26903502

ABSTRACT

During development of the nervous system, growing axons rely on guidance molecules to direct axon pathfinding. A well-characterized family of guidance molecules are the membrane-associated ephrins, which together with their cognate Eph receptors, direct axon navigation in a contact-mediated fashion. InC. elegans, the ephrin-Eph signaling system is conserved and is best characterized for their roles in neuroblast migration during early embryogenesis. This study demonstrates a role for the C. elegans ephrin EFN-4 in axon guidance. We provide both genetic and biochemical evidence that is consistent with the C. elegans divergent L1 cell adhesion molecule LAD-2 acting as a non-canonical ephrin receptor to EFN-4 to promote axon guidance. We also show that EFN-4 probably functions as a diffusible factor because EFN-4 engineered to be soluble can promote LAD-2-mediated axon guidance. This study thus reveals a potential additional mechanism for ephrins in regulating axon guidance and expands the repertoire of receptors by which ephrins can signal.


Subject(s)
Axons/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/embryology , Ephrins/genetics , Nervous System/embryology , Neural Cell Adhesion Molecule L1/genetics , Neurogenesis/physiology , Animals , Cell Line , HEK293 Cells , Humans , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Morphogenesis , Receptors, Eph Family/genetics , Signal Transduction
4.
Genetics ; 199(2): 497-509, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25488979

ABSTRACT

The L1CAM family of cell adhesion molecules is a conserved set of single-pass transmembrane proteins that play diverse roles required for proper nervous system development and function. Mutations in L1CAMs can cause the neurological L1 syndrome and are associated with autism and neuropsychiatric disorders. L1CAM expression in the mature nervous system suggests additional functions besides the well-characterized developmental roles. In this study, we demonstrate that the gene encoding the Caenorhabditis elegans L1CAM, sax-7, genetically interacts with gtl-2, as well as with unc-13 and rab-3, genes that function in neurotransmission. These sax-7 genetic interactions result in synthetic phenotypes that are consistent with abnormal synaptic function. Using an inducible sax-7 expression system and pharmacological reagents that interfere with cholinergic transmission, we uncovered a previously uncharacterized nondevelopmental role for sax-7 that impinges on synaptic function.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Synapses/metabolism , Aldicarb/pharmacology , Animals , Animals, Genetically Modified , Culture Media , Epistasis, Genetic , Gene Expression , Mutation , Neurons/drug effects , Neurons/metabolism , Organ Specificity/drug effects , Organ Specificity/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
5.
Dev Biol ; 398(2): 255-66, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25523392

ABSTRACT

Caenorhabditis elegans molting is a process during which the apical extracellular matrix of the epidermis, the cuticle, is remodeled through a process of degradation and re-synthesis. Using a genetic approach, we identified nekl-3 as essential for the completion of molting. NEKL-3 is highly similar to the mammalian NEK kinase family members NEK6 and NEK7. Animals homozygous for a hypomorphic mutation in nekl-3, sv3, had a novel molting defect in which the central body region, but not the head or tail, was unable to shed the old cuticle. In contrast, a null mutation in nekl-3, gk506, led to complete enclosure within the old cuticle. nekl-2, which is most similar to mammalian NEK8, was also essential for molting. Mosaic analyses demonstrated that NEKL-2 and NEKL-3 were specifically required within the large epidermal syncytium, hyp7, to facilitate molting. Consistent with this, NEKL-2 and NEKL-3 were expressed at the apical surface of hyp7 where they localized to small spheres or tubular structures. Inhibition of nekl-2, but not nekl-3, led to the mislocalization of LRP-1/megalin, a cell surface receptor for low-density lipoprotein (LDL)-binding proteins. In addition, nekl-2 inhibition led to the mislocalization of several other endosome-associated proteins. Notably, LRP-1 acts within hyp7 to facilitate completion of molting, suggesting at least one mechanism by which NEKL-2 may influence molting. Notably, our studies failed to reveal a requirement for NEKL-2 or NEKL-3 in cell division, a function reported for several mammalian NEKs including NEK6 and NEK7. Our findings provide the first genetic and in vivo evidence for a role of NEK family members in endocytosis, which may be evolutionarily conserved.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/physiology , Molting , Protein Kinases/metabolism , Alleles , Amino Acid Sequence , Animals , Biomarkers/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Polarity , Endocytosis , Endosomes/metabolism , Fluorescence , Genes, Helminth , Genes, Reporter , Green Fluorescent Proteins/metabolism , Molecular Sequence Data , Mosaicism , Mutation/genetics , NIMA-Related Kinases , Phenotype , Phylogeny , Protein Kinases/chemistry , Protein Kinases/genetics , Sequence Alignment , Subcutaneous Tissue/embryology , Subcutaneous Tissue/metabolism
6.
Mol Biol Cell ; 24(3): 308-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23242996

ABSTRACT

Low-density lipoprotein receptor (LDLR) internalization clears cholesterol-laden LDL particles from circulation in humans. Defects in clathrin-dependent LDLR endocytosis promote elevated serum cholesterol levels and can lead to atherosclerosis. However, our understanding of the mechanisms that control LDLR uptake remains incomplete. To identify factors critical to LDLR uptake, we pursued a genome-wide RNA interference screen using Caenorhabditis elegans LRP-1/megalin as a model for LDLR transport. In doing so, we discovered an unanticipated requirement for the clathrin-binding endocytic adaptor epsin1 in LDLR endocytosis. Epsin1 depletion reduced LDLR internalization rates in mammalian cells, similar to the reduction observed following clathrin depletion. Genetic and biochemical analyses of epsin in C. elegans and mammalian cells uncovered a requirement for the ubiquitin-interaction motif (UIM) as critical for receptor transport. As the epsin UIM promotes the internalization of some ubiquitinated receptors, we predicted LDLR ubiquitination as necessary for endocytosis. However, engineered ubiquitination-impaired LDLR mutants showed modest internalization defects that were further enhanced with epsin1 depletion, demonstrating epsin1-mediated LDLR endocytosis is independent of receptor ubiquitination. Finally, we provide evidence that epsin1-mediated LDLR uptake occurs independently of either of the two documented internalization motifs (FxNPxY or HIC) encoded within the LDLR cytoplasmic tail, indicating an additional internalization mechanism for LDLR.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Caenorhabditis elegans/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Amino Acid Substitution , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Endocytosis , Gene Knockdown Techniques , HeLa Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Molecular Sequence Data , Protein Stability , Protein Transport , RNA Interference , Ubiquitination
7.
Curr Biol ; 21(10): 883-8, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21549603

ABSTRACT

Neuronal networks operate over a wide range of activity levels, with both neuronal and nonneuronal cells contributing to the balance of excitation and inhibition. Activity imbalance within neuronal networks underlies many neurological diseases, such as epilepsy. The Caenorhabditis elegans locomotor circuit operates via coordinated activity of cholinergic excitatory and GABAergic inhibitory transmission. We have previously shown that a gain-of-function mutation in a neuronal acetylcholine receptor, acr-2(gf), causes an epileptic-like convulsion behavior. Here we report that the behavioral and physiological effects of acr-2(gf) require the activity of the TRPM channel GTL-2 in nonneuronal tissues. Loss of gtl-2 function does not affect baseline synaptic transmission but can compensate for the excitation-inhibition imbalance caused by acr-2(gf). The compensatory effects of removing gtl-2 are counterbalanced by another TRPM channel, GTL-1, and can be recapitulated by acute treatment with divalent cation chelators, including those specific for Zn(2+). Together, these data reveal an important role for ion homeostasis in the balance of neuronal network activity and a novel function of nonneuronal TRPM channels in the fine-tuning of this network activity.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Homeostasis/physiology , Ions/metabolism , Receptors, Nicotinic/metabolism , Seizures/metabolism , TRPM Cation Channels/genetics , Amino Acid Sequence , Animals , Caenorhabditis elegans , Electrophysiology , Larva/physiology , Magnesium , Microscopy, Fluorescence , Molecular Sequence Data , Mutation/genetics , Polymorphism, Single Nucleotide , RNA Interference , Receptors, Nicotinic/genetics , Sequence Analysis, DNA
8.
J Cell Biol ; 192(2): 349-63, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21242290

ABSTRACT

The dystrophin protein complex (DPC), composed of dystrophin and associated proteins, is essential for maintaining muscle membrane integrity. The link between mutations in dystrophin and the devastating muscle failure of Duchenne's muscular dystrophy (DMD) has been well established. Less well appreciated are the accompanying cognitive impairment and neuropsychiatric disorders also presented in many DMD patients, which suggest a wider role for dystrophin in membrane-cytoskeleton function. This study provides genetic evidence of a novel role for DYS-1/dystrophin in maintaining neural organization in Caenorhabditis elegans. This neuronal function is distinct from the established role of DYS-1/dystrophin in maintaining muscle integrity and regulating locomotion. SAX-7, an L1 cell adhesion molecule (CAM) homologue, and STN-2/γ-syntrophin also function to maintain neural integrity in C. elegans. This study provides biochemical data that show that SAX-7 associates with DYS-1 in an STN-2/γ-syntrophin-dependent manner. These results reveal a recruitment of L1CAMs to the DPC to ensure neural integrity is maintained.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Dystrophin/metabolism , Neurons/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Cells, Cultured , Cytoskeleton/metabolism , Dystrophin/genetics , Dystrophin-Associated Proteins/metabolism , Humans , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/cytology , Protein Binding , Two-Hybrid System Techniques
9.
Dev Dyn ; 239(5): 1490-501, 2010 May.
Article in English | MEDLINE | ID: mdl-20225255

ABSTRACT

The L1 family of cell adhesion molecules (L1CAMs) in vertebrates has long been studied for its roles in nervous system development and function. Members of this family have been associated with distinct neurological disorders that include CRASH, autism, 3p syndrome, and schizophrenia. The conservation of L1CAMs in Drosophila and Caenorhabditis elegans allows the opportunity to take advantage of these simple model organisms and their accessible genetic manipulations to dissect L1CAM functions and mechanisms of action. This review summarizes the discoveries of L1CAMs made in C. elegans, showcasing this simple model organism as a powerful system to uncover L1CAM mechanisms and roles in healthy and diseased states.


Subject(s)
Neural Cell Adhesion Molecule L1/physiology , Animals , Caenorhabditis elegans , Nervous System/growth & development , Nervous System Diseases
10.
Genetics ; 180(3): 1429-43, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18791240

ABSTRACT

The L1 family of single-pass transmembrane cell adhesion molecules (L1CAMs) is conserved from Caenorhabditis elegans and Drosophila to vertebrates and is required for axon guidance, neurite outgrowth, and maintenance of neuronal positions. The extracellular region of L1CAMs mediates cell adhesion via interactions with diverse cell-surface and extracellular matrix proteins. In contrast, less is known regarding the function of the intracellular domains in the L1CAM cytoplasmic tail. Previously, we identified a role of the C. elegans L1CAM homolog, SAX-7, in maintaining neuronal and axonal positioning. Here, we demonstrate that this function is dependent on three conserved motifs that reside in the SAX-7 cytoplasmic tail: (1) the FERM-binding motif, (2) the ankyrin-binding domain, and (3) the PDZ-binding motif. Furthermore, we provide molecular and genetic evidence that UNC-44 ankyrin and STN-2 gamma-syntrophin bind SAX-7 via the respective ankyrin-binding and PDZ-binding motifs to regulate SAX-7 function in maintaining neuronal positioning.


Subject(s)
Ankyrins/metabolism , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/physiology , Neural Cell Adhesion Molecules/genetics , Neurons/physiology , Animals , Animals, Genetically Modified , Ankyrins/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Humans , Kidney/metabolism , Muscle Proteins/genetics , Muscle, Striated/physiology , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/metabolism , Neurons/cytology , Phenotype , Sarcomeres/physiology , Two-Hybrid System Techniques
11.
J Cell Biol ; 180(1): 233-46, 2008 Jan 14.
Article in English | MEDLINE | ID: mdl-18195110

ABSTRACT

The L1 cell adhesion molecule (L1CAM) participates in neuronal development. Mutations in the human L1 gene can cause the neurological disorder CRASH (corpus callosum hypoplasia, retardation, adducted thumbs, spastic paraplegia, and hydrocephalus). This study presents genetic data that shows that L1-like adhesion gene 2 (LAD-2), a Caenorhabditis elegans L1CAM, functions in axon pathfinding. In the SDQL neuron, LAD-2 mediates dorsal axon guidance via the secreted MAB-20/Sema2 and PLX-2 plexin receptor, the functions of which have largely been characterized in epidermal morphogenesis. We use targeted misexpression experiments to provide in vivo evidence that MAB-20/Sema2 acts as a repellent to SDQL. Coimmunoprecipitation assays reveal that MAB-20 weakly interacts with PLX-2; this interaction is increased in the presence of LAD-2, which can interact independently with MAB-20 and PLX-2. These results suggest that LAD-2 functions as a MAB-20 coreceptor to secure MAB-20 coupling to PLX-2. In vertebrates, L1 binds neuropilin1, the obligate receptor to the secreted Sema3A. However, invertebrates lack neuropilins. LAD-2 may thus function in the semaphorin complex by combining the roles of neuropilins and L1CAMs.


Subject(s)
Axons/metabolism , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Membrane Proteins/physiology , Molecular Sequence Data , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecules/metabolism , Receptors, Cell Surface/metabolism , Sequence Alignment , Signal Transduction
12.
Dev Biol ; 284(2): 273-91, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16023097

ABSTRACT

The L1 family of cell adhesion molecules (L1CAMs) is important for neural development. Mutations in one of the human L1CAM genes, L1, can result in several neurological syndromes, the symptoms of which are variably penetrant. The physiological cause of these symptoms, collectively termed CRASH, is not clear. Caenorhabditis elegans animals genetically null for the L1CAM homologue LAD-1, exhibit variably penetrant pleiotropic phenotypes that are similar to the CRASH symptoms; thus the C. elegans lad-1 mutant provides an excellent model system to study how disruption of L1 leads to these abnormalities. These phenotypes include uncoordinated movements, variable embryonic lethality, and abnormal neuronal distribution and axon trajectories. Our analysis revealed that many of these phenotypes are likely a result of tissue detachment.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion , Helminth Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Animals, Genetically Modified , Axons , Caenorhabditis elegans/cytology , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Adhesion Molecules, Neuronal/genetics , Chromosome Mapping , Genes, Helminth , Genes, Reporter , Genetic Complementation Test , Green Fluorescent Proteins/metabolism , Helminth Proteins/genetics , Microscopy, Video , Models, Biological , Morphogenesis , Muscles/embryology , Muscles/metabolism , Mutation , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL