Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Med Genet ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670634

ABSTRACT

BACKGROUND: Pontocerebellar hypoplasia (PCH) may present with supratentorial phenotypes and is often accompanied by microcephaly. Damaging mutations in the X-linked gene CASK produce self-limiting microcephaly with PCH in females but are often lethal in males. CASK deficiency leads to early degeneration of cerebellar granule cells but its role in other regions of the brain remains uncertain. METHOD: We generated a conditional Cask knockout mice and deleted Cask ubiquitously after birth at different times. We examined the clinical features in several subjects with damaging mutations clustered in the central part of the CASK protein. We have performed phylogenetic analysis and RT-PCR to assess the splicing pattern within the same protein region and performed in silico structural analysis to examine the effect of splicing on the CASK's structure. RESULT: We demonstrate that deletion of murine Cask after adulthood does not affect survival but leads to cerebellar degeneration and ataxia over time. Intriguingly, damaging hemizygous CASK mutations in boys who display microcephaly and cerebral dysfunction but without PCH are known. These mutations are present in two vertebrate-specific CASK exons. These exons are subject to alternative splicing both in forebrain and hindbrain. Inclusion of these exons differentially affects the molecular structure and hence possibly the function/s of the CASK C-terminus. CONCLUSION: Loss of CASK function disproportionately affects the cerebellum. Clinical data, however, suggest that CASK may have additional vertebrate-specific function/s that play a role in the mammalian forebrain. Thus, CASK has an ancient function shared between invertebrates and vertebrates as well as novel vertebrate-specific function/s.

2.
Cell ; 133(2): 328-39, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423203

ABSTRACT

CASK is a unique MAGUK protein that contains an N-terminal CaM-kinase domain besides the typical MAGUK domains. The CASK CaM-kinase domain is presumed to be a catalytically inactive pseudokinase because it lacks the canonical DFG motif required for Mg2+ binding that is thought to be indispensable for kinase activity. Here we show, however, that CASK functions as an active protein kinase even without Mg2+ binding. High-resolution crystal structures reveal that the CASK CaM-kinase domain adopts a constitutively active conformation that binds ATP and catalyzes phosphotransfer without Mg2+. The CASK CaM-kinase domain phosphorylates itself and at least one physiological interactor, the synaptic protein neurexin-1, to which CASK is recruited via its PDZ domain. Thus, our data indicate that CASK combines the scaffolding activity of MAGUKs with an unusual kinase activity that phosphorylates substrates recuited by the scaffolding activity. Moreover, our study suggests that other pseudokinases (10% of the kinome) could also be catalytically active.


Subject(s)
Glycoproteins/metabolism , Guanylate Kinases/chemistry , Guanylate Kinases/metabolism , Neuropeptides/metabolism , Animals , Cell Line , Cells, Cultured , Crystallography, X-Ray , Humans , Magnesium/metabolism , Mice , Models, Molecular , Neurons/metabolism , Nucleotides/metabolism , Protein Binding , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
3.
J Med Genet ; 59(11): 1044-1057, 2022 11.
Article in English | MEDLINE | ID: mdl-35149592

ABSTRACT

BACKGROUND: Heterozygous loss of X-linked genes like CASK and MeCP2 (Rett syndrome) causes developmental delay in girls, while in boys, loss of the only allele of these genes leads to epileptic encephalopathy. The mechanism for these disorders remains unknown. CASK-linked cerebellar hypoplasia is presumed to result from defects in Tbr1-reelin-mediated neuronal migration. METHOD: Here we report clinical and histopathological analyses of a deceased 2-month-old boy with a CASK-null mutation. We next generated a mouse line where CASK is completely deleted (hemizygous and homozygous) from postmigratory neurons in the cerebellum. RESULT: The CASK-null human brain was smaller in size but exhibited normal lamination without defective neuronal differentiation, migration or axonal guidance. The hypoplastic cerebellum instead displayed astrogliosis and microgliosis, which are markers for neuronal loss. We therefore hypothesise that CASK loss-induced cerebellar hypoplasia is the result of early neurodegeneration. Data from the murine model confirmed that in CASK loss, a small cerebellum results from postdevelopmental degeneration of cerebellar granule neurons. Furthermore, at least in the cerebellum, functional loss from CASK deletion is secondary to degeneration of granule cells and not due to an acute molecular functional loss of CASK. Intriguingly, female mice with heterozygous deletion of CASK in the cerebellum do not display neurodegeneration. CONCLUSION: We suggest that X-linked neurodevelopmental disorders like CASK mutation and Rett syndrome are pathologically neurodegenerative; random X-chromosome inactivation in heterozygous mutant girls, however, results in 50% of cells expressing the functional gene, resulting in a non-progressive pathology, whereas complete loss of the only allele in boys leads to unconstrained degeneration and encephalopathy.


Subject(s)
Cerebellar Diseases , Neurodegenerative Diseases , Rett Syndrome , Male , Humans , Animals , Female , Mice , Infant , Genes, X-Linked/genetics , Guanylate Kinases/genetics , Rett Syndrome/genetics , Cerebellar Diseases/genetics , Neurodegenerative Diseases/genetics
4.
Eur J Neurosci ; 54(6): 5982-5999, 2021 09.
Article in English | MEDLINE | ID: mdl-34449932

ABSTRACT

Neuropathy, or dysfunction of peripheral nerve, is one of the most common neurological manifestation in patients with diabetes mellitus (DM). DM is typically associated with a hyperglycaemic milieu, which promotes non-enzymatic glycation of proteins. Proteins with advanced glycation are known to engage a cell-surface receptor called the receptor for advanced glycation end products (RAGE). Thus, it is reasonable to assume that RAGE and its associated molecule-mediated cellular signalling may contribute to DM-induced symmetrical axonal (length-dependent) neuropathy. Of particular interest is diaphanous related formin 1 (DIAPH1), a cytoskeletal organizing molecule, which interacts with the cytosolic domain of RAGE and whose dysfunction may precipitate axonopathy/neuropathy. Indeed, it has been demonstrated that both RAGE and DIAPH1 are expressed in the motor and sensory fibres of nerve harvested from DM animal models. Although the detailed molecular role of RAGE and DIAPH1 in diabetic neurological complications remains unclear, here we will discuss available evidence of their involvement in peripheral diabetic neuropathy. Specifically, we will discuss how a hyperglycaemic environment is not only likely to elevate advanced glycation end products (ligands of RAGE) and induce a pro-inflammatory environment but also alter signalling via RAGE and DIAPH1. Further, hyperglycaemia may regulate epigenetic mechanisms that interacts with RAGE signalling. We suggest the cumulative effect of hyperglycaemia on RAGE-DIAPH1-mediated signalling may be disruptive to axonal cytoskeletal organization and transport and is therefore likely to play a key role in pathogenesis of diabetic symmetrical axonal neuropathy.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Diabetic Neuropathies , Animals , Formins , Humans , Ligands , Receptor for Advanced Glycation End Products , Signal Transduction
5.
Biochem Biophys Res Commun ; 559: 92-98, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33933994

ABSTRACT

Synaptic transmission is mediated by neurotransmitters that are stored in synaptic vesicles (SV) and released at the synaptic active zone (AZ). While in recent years major progress has been made in unraveling the molecular machinery responsible for SV docking, fusion and exocytosis, the mechanisms governing AZ protein and SV trafficking through axons still remain unclear. Here, we performed stop-flow nerve ligation to examine axonal trafficking of endogenous AZ and SV proteins. Rat sciatic nerves were collected 1 h, 3 h and 8 h post ligation and processed for immunohistochemistry and electron microscopy. First, we followed the transport of an integral synaptic vesicle protein, SV2A and a SV-associated protein involved in SV trafficking, Rab3a, and observed that while SV2A accumulated on both sides of ligation, Rab3a was only noticeable in the proximal segment of the ligated nerve indicating that only SV trans-membrane protein SV2A displayed a bi-directional axonal transport. We then demonstrate that multiple AZ proteins accumulate rapidly on either side of the ligation with a timescale similar to that of SV2A. Overall, our data uncovers an unexpected robust bi-directional, coordinated -trafficking of SV and AZ proteins in peripheral nerves. This implies that pathological disruption of axonal trafficking will not only impair trafficking of newly synthesized proteins to the synapse but will also affect retrograde transport, leading to neuronal dysfunction and likely neurodegeneration.


Subject(s)
Axonal Transport , Nerve Tissue Proteins/metabolism , Peripheral Nerves/physiology , Protein Transport , Synaptic Vesicles/metabolism , Animals , Male , Rats, Sprague-Dawley , Synaptic Transmission
6.
Am J Med Genet A ; 179(1): 94-103, 2019 01.
Article in English | MEDLINE | ID: mdl-30549415

ABSTRACT

Heterozygous loss-of-function mutations in the X-linked gene CASK are associated with mental retardation and microcephaly with pontine and cerebellar hypoplasia (MICPCH) and ophthalmological disorders including optic nerve atrophy (ONA) and optic nerve hypoplasia (ONH). Recently, we have demonstrated that CASK(+/-) mice display ONH with 100% penetrance but exhibit no change in retinal lamination or structure. It is not clear if CASK loss-of-function predominantly affects retinal ganglion cells, or if other retinal cells like photoreceptors are also involved. Here, we report a heterozygous missense mutation in the N-terminal calcium/calmodulin-dependent kinase (CaMK) domain of the CASK protein in which a highly conserved leucine is mutated to the cyclic amino acid proline. In silico analysis suggests that the mutation may produce destabilizing structural changes. Experimentally, we observe pronounced misfolding and insolubility of the CASKL209P protein. Interestingly, the remaining soluble mutant protein fails to interact with Mint1, which specifically binds to CASK's CaMK domain, suggesting a mechanism for the phenotypes observed with the CASKL209P mutation. In addition to microcephaly, cerebellar hypoplasia and delayed development, the subject with the L209P mutation also presented with bilateral retinal dystrophy and ONA. Electroretinography indicated that rod photoreceptors are the most prominently affected cells. Our data suggest that the CASK interactions mediated by the CaMK domain may play a crucial role in retinal function, and thus, in addition to ONH, individuals with mutations in the CASK gene may exhibit other retinal disorders, depending on the nature of mutation.


Subject(s)
Atrophy/genetics , Guanylate Kinases/genetics , Microcephaly/genetics , Retinal Dystrophies/genetics , Adaptor Proteins, Signal Transducing/genetics , Atrophy/diagnostic imaging , Atrophy/physiopathology , Child , Female , Guanylate Kinases/chemistry , HEK293 Cells , Heterozygote , Humans , Loss of Function Mutation/genetics , Microcephaly/diagnostic imaging , Microcephaly/physiopathology , Molecular Dynamics Simulation , Mutation, Missense/genetics , Nerve Tissue Proteins/genetics , Optic Nerve/physiopathology , Photoreceptor Cells/pathology , Protein Folding , Retinal Dystrophies/diagnostic imaging , Retinal Dystrophies/physiopathology , Retinal Ganglion Cells/pathology , Exome Sequencing
7.
Hum Genet ; 137(3): 231-246, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29426960

ABSTRACT

Deletion and truncation mutations in the X-linked gene CASK are associated with severe intellectual disability (ID), microcephaly and pontine and cerebellar hypoplasia in girls (MICPCH). The molecular origin of CASK-linked MICPCH is presumed to be due to disruption of the CASK-Tbr-1 interaction. This hypothesis, however, has not been directly tested. Missense variants in CASK are typically asymptomatic in girls. We report three severely affected girls with heterozygous CASK missense mutations (M519T (2), G659D (1)) who exhibit ID, microcephaly, and hindbrain hypoplasia. The mutation M519T results in the replacement of an evolutionarily invariant methionine located in the PDZ signaling domain known to be critical for the CASK-neurexin interaction. CASKM519T is incapable of binding to neurexin, suggesting a critically important role for the CASK-neurexin interaction. The mutation G659D is in the SH3 (Src homology 3) domain of CASK, replacing a semi-conserved glycine with aspartate. We demonstrate that the CASKG659D mutation affects the CASK protein in two independent ways: (1) it increases the protein's propensity to aggregate; and (2) it disrupts the interface between CASK's PDZ (PSD95, Dlg, ZO-1) and SH3 domains, inhibiting the CASK-neurexin interaction despite residing outside of the domain deemed critical for neurexin interaction. Since heterozygosity of other aggregation-inducing mutations (e.g., CASKW919R) does not produce MICPCH, we suggest that the G659D mutation produces microcephaly by disrupting the CASK-neurexin interaction. Our results suggest that disruption of the CASK-neurexin interaction, not the CASK-Tbr-1 interaction, produces microcephaly and cerebellar hypoplasia. These findings underscore the importance of functional validation for variant classification.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cerebellum/abnormalities , Genetic Diseases, X-Linked/genetics , Guanylate Kinases/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Nervous System Malformations/genetics , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/chemistry , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Genetic Diseases, X-Linked/physiopathology , Guanylate Kinases/chemistry , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Microcephaly/diagnostic imaging , Microcephaly/physiopathology , Mutation, Missense/genetics , Nerve Tissue Proteins/chemistry , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/physiopathology , Neural Cell Adhesion Molecules , PDZ Domains/genetics , Phenotype , Protein Aggregates/genetics , Protein Binding , Protein Interaction Maps/genetics , T-Box Domain Proteins/genetics , src Homology Domains/genetics
8.
Cell Mol Life Sci ; 73(18): 3599-621, 2016 09.
Article in English | MEDLINE | ID: mdl-27015872

ABSTRACT

CASK, a MAGUK family protein, is an essential protein present in the presynaptic compartment. CASK's cellular role is unknown, but it interacts with multiple proteins important for synapse formation and function, including neurexin, liprin-α, and Mint1. CASK phosphorylates neurexin in a divalent ion-sensitive manner, although the functional relevance of this activity is unclear. Here we find that liprin-α and Mint1 compete for direct binding to CASK, but neurexin1ß eliminates this competition, and all four proteins form a complex. We describe a novel mode of interaction between liprin-α and CASK when CASK is bound to neurexin1ß. We show that CASK phosphorylates neurexin, modulating the interaction of liprin-α with the CASK-neurexin1ß-Mint1 complex. Thus, CASK creates a regulatory and structural link between the presynaptic adhesion molecule neurexin and active zone organizer, liprin-α. In neuronal culture, CASK appears to regulate the stability of neurexin by linking it with this multi-protein presynaptic active zone complex.


Subject(s)
Guanylate Kinases/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Cell Membrane/metabolism , Cells, Cultured , Female , Guanylate Kinases/chemistry , Guanylate Kinases/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/chemistry , Neurons/cytology , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment
9.
Proc Natl Acad Sci U S A ; 110(30): E2812-20, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23821748

ABSTRACT

Membrane fusion is mediated by complexes formed by SNAP-receptor (SNARE) and Secretory 1 (Sec1)/mammalian uncoordinated-18 (Munc18)-like (SM) proteins, but it is unclear when and how these complexes assemble. Here we describe an improved two-color fluorescence nanoscopy technique that can achieve effective resolutions of up to 7.5-nm full width at half maximum (3.2-nm localization precision), limited only by stochastic photon emission from single molecules. We use this technique to dissect the spatial relationships between the neuronal SM protein Munc18-1 and SNARE proteins syntaxin-1 and SNAP-25 (25 kDa synaptosome-associated protein). Strikingly, we observed nanoscale clusters consisting of syntaxin-1 and SNAP-25 that contained associated Munc18-1. Rescue experiments with syntaxin-1 mutants revealed that Munc18-1 recruitment to the plasma membrane depends on the Munc18-1 binding to the N-terminal peptide of syntaxin-1. Our results suggest that in a primary neuron, SNARE/SM protein complexes containing syntaxin-1, SNAP-25, and Munc18-1 are preassembled in microdomains on the presynaptic plasma membrane. Our superresolution imaging method provides a framework for investigating interactions between the synaptic vesicle fusion machinery and other subcellular systems in situ.


Subject(s)
Microscopy, Fluorescence/methods , Munc18 Proteins/chemistry , SNARE Proteins/chemistry , Synaptosomal-Associated Protein 25/chemistry , Syntaxin 1/chemistry
11.
Biochem Soc Trans ; 41(4): 1017-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863172

ABSTRACT

CASK (Ca2+/calmodulin-activated serine kinase) is a synaptic protein that interacts with the cytosolic tail of adhesion molecules such as neurexins, syncam and syndecans. It belongs to the MAGUK (membrane-associated guanylate kinase) family of scaffolding proteins which are known to decorate cell-cell junctions. CASK is an essential gene in mammals, critical for neurodevelopment. Mutations in the CASK gene in humans result in phenotypes that range from intellectual disability to lethality. Despite its importance, CASK has a single genetic isoform located in the short arm of the X chromosome near an evolutionary breakpoint. Surprisingly, CASK is a non-essential gene in invertebrates and displays functional divergence. In the present article, we describe the phylogenetic differences in existing CASK orthologues. The CASK gene has undergone a huge expansion in size (~55-fold). Almost all of this expansion is a direct result of an increase in the size of the introns. The coding region of CASK orthologues, and hence the protein, exhibit a high degree of evolutionary conservation. Within the protein, domain arrangement is completely conserved and substitution rates are higher in the connecting loop regions [L27 (Lin2, Lin7)] than within the domain. Our analyses of single residue substitutions and genotype-phenotype relationships suggest that, other than intronic expansion, the dramatic functional changes of CASK are driven by subtle (non-radical) primary structure changes within the CASK protein and concomitant changes in its protein interactors.


Subject(s)
Guanylate Kinases/chemistry , Animals , Chromosomes, Human, X , Evolution, Molecular , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Humans , Introns , Models, Molecular , Phylogeny , Protein Conformation
12.
Proc Natl Acad Sci U S A ; 107(14): 6504-9, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20332206

ABSTRACT

Piccolo and bassoon are highly homologous multidomain proteins of the presynaptic cytomatrix whose function is unclear. Here, we generated piccolo knockin/knockout mice that either contain wild-type levels of mutant piccolo unable to bind Ca(2+) (knockin), approximately 60% decreased levels of piccolo that is C-terminally truncated (partial knockout), or <5% levels of piccolo (knockout). All piccolo mutant mice were viable and fertile, but piccolo knockout mice exhibited increased postnatal mortality. Unexpectedly, electrophysiology and electron microscopy of piccolo-deficient synapses failed to uncover a major phenotype either in acute hippocampal slices or in cultured cortical neurons. To unmask potentially redundant functions of piccolo and bassoon, we thus acutely knocked down expression of bassoon in wild-type and piccolo knockout neurons. Despite a nearly complete loss of piccolo and bassoon, however, we still did not detect an electrophysiological phenotype in cultured piccolo- and bassoon-deficient neurons in either GABAergic or glutamatergic synaptic transmission. In contrast, electron microscopy revealed a significant reduction in synaptic vesicle clustering in double bassoon/piccolo-deficient synapses. Thus, we propose that piccolo and bassoon play a redundant role in synaptic vesicle clustering in nerve terminals without directly participating in neurotransmitter release.


Subject(s)
Cytoskeletal Proteins/metabolism , Exocytosis , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuropeptides/metabolism , Synaptic Vesicles/metabolism , Amino Acid Sequence , Animals , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neuropeptides/chemistry , Neuropeptides/genetics , Synaptic Transmission , Synaptic Vesicles/ultrastructure
13.
J Mol Med (Berl) ; 101(8): 1015-1028, 2023 08.
Article in English | MEDLINE | ID: mdl-37462767

ABSTRACT

Multiple molecular pathways including the receptor for advanced glycation end-products-diaphanous related formin 1 (RAGE-Diaph1) signaling are known to play a role in diabetic peripheral neuropathy (DPN). Evidence suggests that neuropathological alterations in type 1 diabetic spinal cord may occur at the same time as or following peripheral nerve abnormalities. We demonstrated that DPN was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. More than 500 differentially expressed genes (DEGs) belonging to multiple functional pathways were identified in diabetic spinal cord and of those the most enriched was RAGE-Diaph1 related PI3K-Akt pathway. Only seven of spinal cord DEGs overlapped with DEGs from type 1 diabetic sciatic nerve and only a single gene cathepsin E (CTSE) was common for both type 1 and type 2 diabetic mice. In silico analysis suggests that molecular changes in spinal cord may act synergistically with RAGE-Diaph1 signaling axis in the peripheral nerve. KEY MESSAGES: Molecular perturbations in spinal cord may be involved in the progression of diabetic peripheral neuropathy. Diabetic peripheral neuropathy was associated with perturbations of RAGE-Diaph1 signaling pathway in peripheral nerve accompanied by widespread spinal cord molecular changes. In silico analysis revealed that PI3K-Akt signaling axis related to RAGE-Diaph1 was the most enriched biological pathway in diabetic spinal cord. Cathepsin E may be the target molecular hub for intervention against diabetic peripheral neuropathy.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetic Neuropathies , Hyperglycemia , Animals , Mice , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/complications , Cathepsin E , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sciatic Nerve/pathology , Hyperglycemia/genetics , Hyperglycemia/pathology
14.
Cells ; 11(7)2022 03 28.
Article in English | MEDLINE | ID: mdl-35406695

ABSTRACT

Most human disease manifests as a result of tissue pathology, due to an underlying disease process (pathogenesis), rather than the acute loss of specific molecular function(s). Successful therapeutic strategies thus may either target the correction of a specific molecular function or halt the disease process. For the vast majority of brain diseases, clear etiologic and pathogenic mechanisms are still elusive, impeding the discovery or design of effective disease-modifying drugs. The development of valid animal models and their proper characterization is thus critical for uncovering the molecular basis of the underlying pathobiological processes of brain disorders. MICPCH (microcephaly and pontocerebellar hypoplasia) is a monogenic condition that results from variants of an X-linked gene, CASK (calcium/calmodulin-dependent serine protein kinase). CASK variants are associated with a wide range of clinical presentations, from lethality and epileptic encephalopathies to intellectual disabilities, microcephaly, and autistic traits. We have examined CASK loss-of-function mutations in model organisms to simultaneously understand the pathogenesis of MICPCH and the molecular function/s of CASK. Our studies point to a highly complex relationship between the potential molecular function/s of CASK and the phenotypes observed in model organisms and humans. Here we discuss the implications of our observations from the pathogenesis of MICPCH as a cautionary narrative against oversimplifying molecular interpretations of data obtained from genetically modified animal models of human diseases.


Subject(s)
Mental Retardation, X-Linked , Microcephaly , Nervous System Malformations , Animals , Guanylate Kinases/genetics , Mental Retardation, X-Linked/complications , Mice , Microcephaly/genetics , Nervous System Malformations/genetics
15.
Neurosci Bull ; 38(10): 1248-1262, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35729453

ABSTRACT

This review reflects upon our own as well as other investigators' studies on the role of receptor for advanced glycation end-products (RAGE), bringing up the latest information on RAGE in physiology and pathology of the nervous system. Over the last ten years, major progress has been made in uncovering many of RAGE-ligand interactions and signaling pathways in nervous tissue; however, the translation of these discoveries into clinical practice has not come to fruition yet. This is likely, in part to be the result of our incomplete understanding of this crucial signaling pathway. Clinical trials examining the therapeutic efficacy of blocking RAGE-external ligand interactions by genetically engineered soluble RAGE or an endogenous RAGE antagonist, has not stood up to its promise; however, other trials with different blocking agents are being considered with hope for therapeutic success in diseases of the nervous system.


Subject(s)
Nervous System Diseases , Humans , Ligands , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/physiology
16.
Mol Genet Genomic Med ; 8(10): e1426, 2020 10.
Article in English | MEDLINE | ID: mdl-32696595

ABSTRACT

BACKGROUND: CASK is an X-linked gene in mammals and its deletion in males is incompatible with life. CASK heterozygous mutations in female patients associate with intellectual disability, microcephaly, pontocerebellar hypoplasia, and optic nerve hypoplasia, whereas CASK hemizygous mutations in males manifest as early infantile epileptic encephalopathy with a grim prognosis. Here, we report a rare case of survival of a male patient harboring a CASK null mutation to adolescent age. METHODS: Trio whole exome sequencing analysis was performed from blood genomic DNA. Magnetic resonance imaging (MRI), magnetic resonance spectroscopy (MRS), and electroencephalogram (EEG) analyses were performed to determine anomalies in brain development, metabolite concentrations, and electrical activity, respectively. RESULTS: Trio-WES analysis identified a de novo c.79C>T (p.Arginine27Ter) mutation in CASK causing a premature translation termination at the very N-terminus of the protein. The 17-years, and 11-month-old male patient displayed profound intellectual disability, microcephaly, dysmorphism, ponto-cerebellar hypoplasia, and intractable epilepsy. His systemic symptoms included overall reduced somatic growth, dysautonomia, ventilator and G tube dependence, and severe osteopenia. Brain MRI revealed a severe cerebellar and brain stem hypoplasia with progressive cerebral atrophy. EEG spectral analysis revealed a global functional defect with generalized background slowing and delta waves dominating even in the awake state. CONCLUSION: This case study is the first to report survival of a male patient carrying a CASK loss-of-function mutation to adolescence and highlights that improved palliative care could extend survival. Moreover, the genomic position encoding Arg27 in CASK may possess an increased susceptibility to mutations.


Subject(s)
Abnormalities, Multiple/genetics , Epilepsy/genetics , Genetic Diseases, X-Linked/genetics , Guanylate Kinases/genetics , Intellectual Disability/genetics , Loss of Function Mutation , Abnormalities, Multiple/pathology , Adolescent , Epilepsy/pathology , Genetic Diseases, X-Linked/pathology , Humans , Intellectual Disability/pathology , Male
17.
Sci Rep ; 9(1): 5752, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30962477

ABSTRACT

Mitochondrial DNA (mtDNA) 3243A > G tRNALeu(UUR) heteroplasmic mutation (m.3243A > G) exhibits clinically heterogeneous phenotypes. While the high mtDNA heteroplasmy exceeding a critical threshold causes mitochondrial encephalomyopathy, lactic acidosis with stroke-like episodes (MELAS) syndrome, the low mtDNA heteroplasmy causes maternally inherited diabetes with or without deafness (MIDD) syndrome. How quantitative differences in mtDNA heteroplasmy produces distinct pathological states has remained elusive. Here we show that despite striking similarities in the energy metabolic gene expression signature, the mitochondrial bioenergetics, biogenesis and fuel catabolic functions are distinct in cells harboring low or high levels of the m.3243 A > G mutation compared to wild type cells. We further demonstrate that the low heteroplasmic mutant cells exhibit a coordinate induction of transcriptional regulators of the mitochondrial biogenesis, glucose and fatty acid metabolism pathways that lack in near homoplasmic mutant cells compared to wild type cells. Altogether, these results shed new biological insights on the potential mechanisms by which low mtDNA heteroplasmy may progressively cause diabetes mellitus.


Subject(s)
DNA, Mitochondrial/genetics , Energy Metabolism , MELAS Syndrome/genetics , Mutation , Fatty Acids/metabolism , Glucose/metabolism , Humans , Organelle Biogenesis
18.
Invest Ophthalmol Vis Sci ; 60(10): 3584-3594, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31425583

ABSTRACT

Purpose: Heterozygous mutations in the essential X-linked gene CASK associate with optic nerve hypoplasia (ONH) and other retinal disorders in girls. CASK+/- heterozygous knockout mice with mosaic CASK expression exhibit ONH with a loss of retinal ganglion cells (RGCs) but no changes in retinal morphology. It remains unclear if CASK deficiency selectively affects RGCs or also affects other retinal cells. Furthermore, it is not known if CASK expression in RGCs is critical for optic nerve (ON) development and maintenance. Methods: The visual behavior of CASK+/- mice was assessed and electroretinography (ERG) was performed. Using a mouse line with a floxed CASK gene that expresses approximately 40% CASK globally in all cells (hypomorph) under hemizygous and homozygous conditions, we investigated effects of CASK reduction on the retina and ON. CASK then was completely deleted from RGCs to examine its cell-autonomous role. Finally, for the first time to our knowledge, we describe a hemizygous CASK missense mutation in a boy with ONH. Results: CASK+/- heterozygous mutant mice display reduced visual contrast sensitivity, but ERG is indistinguishable from wildtype. CASK hypomorph mice exhibit ONH, but deletion of CASK from RGCs in this background does not exacerbate the condition. The boy with ONH harbors a missense mutation (p.Pro673Leu) that destabilizes CASK and weakens the crucial CASK-neurexin interaction. Conclusions: Our results demonstrate that mosaic or global reduction in CASK expression and/or function disproportionately affects RGCs. CASK expression in RGCs does not appear critical for cell survival, indicating a noncell autonomous role for CASK in the development of ON.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Guanylate Kinases/genetics , Optic Nerve Hypoplasia/genetics , Animals , Cell Survival , Child, Preschool , Contrast Sensitivity/physiology , Electroretinography , Female , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Optic Nerve Hypoplasia/physiopathology , Retina/physiopathology , Retinal Ganglion Cells/enzymology
19.
Matters (Zur) ; 20172017 05 22.
Article in English | MEDLINE | ID: mdl-30148136

ABSTRACT

Scavenger receptors (SRs) are a family of receptors displaying affinity for a wide variety of ligands including modified lipoproteins. SRs may play a range of physiological functions including intracellular transport, lipid transport and pathogen clearance. The role of SRs has been documented in pathologies such as atherosclerosis and Alzheimer's disease. Although most studies on SRs have focused on macrophages, they are also present in other cells like endothelium, smooth muscles and brain tissue. Within brain, due to its functional similarity, SRs have been studied mostly in microglia. However, in situ images from Allen's brain atlas suggest SRs are abundant in neurons. In this study we have used two fluorophore labeled well characterized SR ligand, maleylated-BSA (MBSA) and polyguanylic acid (poloyG) to probe acute cortical slices. Our data indicate that within cortex, neurons avidly endocytose both ligands. Thus in cerebral cortex neurons may have higher number of functional SRs on the surface than other cell-types.

20.
Methods Mol Biol ; 1647: 171-183, 2017.
Article in English | MEDLINE | ID: mdl-28809002

ABSTRACT

Eukaryotic protein kinases are an intensely investigated class of enzymes which have garnered attention due to their usefulness as drug targets. Determining the regulation of ATP binding to a protein kinase is not only critical for understanding function in a cellular context but also for designing kinase-specific molecular inhibitors. Here, we provide a general procedure for characterizing ATP binding to eukaryotic protein kinases. The protocol can be adapted to identify the conditions under which a particular kinase is activated. The approach is simple, requiring only a fluorescent ATP analog such as TNP-ATP or MANT-ATP and an instrument to monitor changes in fluorescence. Although the interaction kinetics between a kinase and a given ATP analog may differ from that of native ATP, this disadvantage is offset by the ease of performing and interpreting this assay. Importantly, it can be optimized to probe a large variety of conditions under which the kinase-nucleotide binding might be affected.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Fluorescent Dyes/chemistry , Protein Kinases/chemistry , ortho-Aminobenzoates/chemistry , Adenosine Triphosphate/chemistry , Eukaryota/enzymology , Guanylate Kinases/chemistry , Guanylate Kinases/metabolism , Kinetics , Protein Kinases/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL