Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 149(7): 1549-64, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726441

ABSTRACT

Secretory fibroblast growth factors (FGFs) and their receptors are known for their regulatory function in the early stages of neural development. FGF13, a nonsecretory protein of the FGF family, is expressed in cerebral cortical neurons during development and is a candidate gene for syndromal and nonspecific forms of X-chromosome-linked mental retardation (XLMR). However, its function during development remains unclear. We show that FGF13 acts intracellularly as a microtubule-stabilizing protein required for axon and leading process development and neuronal migration in the cerebral cortex. FGF13 is enriched in axonal growth cones and interacts directly with microtubules. Furthermore, FGF13 polymerizes tubulins and stabilizes microtubules. The loss of FGF13 impairs neuronal polarization and increases the branching of axons and leading processes. Genetic deletion of FGF13 in mice results in neuronal migration defects in both the neocortex and the hippocampus. FGF13-deficient mice also exhibit weakened learning and memory, which is correlated to XLMR patients' intellectual disability.


Subject(s)
Fibroblast Growth Factors/metabolism , Neurons/cytology , Neurons/metabolism , Amino Acid Sequence , Animals , Axons/metabolism , Cell Movement , Cell Polarity , Cerebral Cortex/metabolism , Disease Models, Animal , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Growth Cones/metabolism , Hippocampus/cytology , Humans , Male , Mental Retardation, X-Linked/metabolism , Mice , Mice, Knockout , Microtubules/metabolism , Molecular Sequence Data , Polymerization , Tubulin/metabolism
2.
EMBO Rep ; 25(3): 1256-1281, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38429579

ABSTRACT

The plant homeodomain zinc-finger protein, PHF6, is a transcriptional regulator, and PHF6 germline mutations cause the X-linked intellectual disability (XLID) Börjeson-Forssman-Lehmann syndrome (BFLS). The mechanisms by which PHF6 regulates transcription and how its mutations cause BFLS remain poorly characterized. Here, we show genome-wide binding of PHF6 in the developing cortex in the vicinity of genes involved in central nervous system development and neurogenesis. Characterization of BFLS mice harbouring PHF6 patient mutations reveals an increase in embryonic neural stem cell (eNSC) self-renewal and a reduction of neural progenitors. We identify a panel of Ephrin receptors (EphRs) as direct transcriptional targets of PHF6. Mechanistically, we show that PHF6 regulation of EphR is impaired in BFLS mice and in conditional Phf6 knock-out mice. Knockdown of EphR-A phenocopies the PHF6 loss-of-function defects in altering eNSCs, and its forced expression rescues defects of BFLS mice-derived eNSCs. Our data indicate that PHF6 directly promotes Ephrin receptor expression to control eNSC behaviour in the developing brain, and that this pathway is impaired in BFLS.


Subject(s)
Epilepsy , Face/abnormalities , Fingers/abnormalities , Growth Disorders , Hypogonadism , Intellectual Disability , Mental Retardation, X-Linked , Obesity , Humans , Mice , Animals , Intellectual Disability/genetics , Repressor Proteins , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Epilepsy/genetics , Epilepsy/metabolism , Transcription Factors
3.
Neurobiol Dis ; 200: 106621, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39097035

ABSTRACT

Allan-Herndon-Dudley syndrome (AHDS) is a rare X-linked disorder that causes severe neurological damage, for which there is no effective treatment. AHDS is due to inactivating mutations in the thyroid hormone transporter MCT8 that impair the entry of thyroid hormones into the brain, resulting in cerebral hypothyroidism. However, the pathophysiology of AHDS is still not fully understood and this is essential to develop therapeutic strategies. Based on evidence suggesting that thyroid hormone deficit leads to alterations in astroglial cells, including gliosis, in this work, we have evaluated astroglial impairments in MCT8 deficiency by means of magnetic resonance imaging, histological, ultrastructural, and immunohistochemical techniques, and by mining available RNA sequencing outputs. Apparent diffusion coefficient (ADC) imaging values obtained from magnetic resonance imaging showed changes indicative of alterations in brain cytoarchitecture in MCT8-deficient patients (n = 11) compared to control subjects (n = 11). Astroglial alterations were confirmed by immunohistochemistry against astroglial markers in autopsy brain samples of an 11-year-old and a 30th gestational week MCT8-deficient subjects in comparison to brain samples from control subjects at similar ages. These findings were validated and further explored in a mouse model of AHDS. Our findings confirm changes in all the astroglial populations of the cerebral cortex in MCT8 deficiency that impact astrocytic metabolic and mitochondrial cellular respiration functions. These impairments arise early in brain development and persist at adult stages, revealing an abnormal distribution, density, morphology of cortical astrocytes, along with altered transcriptome, compatible with an astrogliosis-like phenotype at adult stages. We conclude that astrocytes are potential novel therapeutic targets in AHDS, and we propose ADC imaging as a tool to monitor the progression of neurological impairments and potential effects of treatments in MCT8 deficiency.


Subject(s)
Astrocytes , Brain , Monocarboxylic Acid Transporters , Muscle Hypotonia , Symporters , Thyroid Hormones , Astrocytes/metabolism , Astrocytes/pathology , Animals , Mice , Humans , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Male , Brain/metabolism , Brain/pathology , Thyroid Hormones/metabolism , Child , Symporters/metabolism , Symporters/genetics , Muscle Hypotonia/metabolism , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Female , Adult , Magnetic Resonance Imaging/methods , Muscular Atrophy
4.
Hum Mol Genet ; 30(7): 575-594, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33772537

ABSTRACT

The PHF6 mutation c.1024C > T; p.R342X, is a recurrent cause of Börjeson-Forssman-Lehmann Syndrome (BFLS), a neurodevelopmental disorder characterized by moderate-severe intellectual disability, truncal obesity, gynecomastia, hypogonadism, long tapering fingers and large ears (MIM#301900). Here, we generated transgenic mice with the identical substitution (R342X mice) using CRISPR technology. We show that the p.R342X mutation causes a reduction in PHF6 protein levels, in both human and mice, from nonsense-mediated decay and nonsense-associated alternative splicing, respectively. Magnetic resonance imaging studies indicated that R342X mice had a reduced brain volume on a mixed genetic background but developed hydrocephaly and a high incidence of postnatal death on a C57BL/6 background. Cortical development proceeded normally, while hippocampus and hypothalamus relative brain volumes were altered. A hypoplastic anterior pituitary was also observed that likely contributes to the small size of the R342X mice. Behavior testing demonstrated deficits in associative learning, spatial memory and an anxiolytic phenotype. Taken together, the R342X mice represent a good preclinical model of BFLS that will allow further dissection of PHF6 function and disease pathogenesis.


Subject(s)
Disease Models, Animal , Epilepsy/genetics , Face/abnormalities , Fingers/abnormalities , Genetic Predisposition to Disease/genetics , Growth Disorders/genetics , Hypogonadism/genetics , Mental Retardation, X-Linked/genetics , Mutation , Obesity/genetics , Repressor Proteins/genetics , Animals , Association Learning/physiology , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Cells, Cultured , Epilepsy/metabolism , Epilepsy/physiopathology , Face/physiopathology , Female , Fingers/physiopathology , Gene Expression Profiling/methods , Growth Disorders/metabolism , Growth Disorders/physiopathology , Humans , Hypogonadism/metabolism , Hypogonadism/physiopathology , Magnetic Resonance Imaging/methods , Male , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Obesity/metabolism , Obesity/physiopathology , RNA-Seq/methods , Repressor Proteins/metabolism , Spatial Memory/physiology
5.
Development ; 147(21)2020 10 23.
Article in English | MEDLINE | ID: mdl-32994169

ABSTRACT

Börjeson-Forssman-Lehmann syndrome (BFLS) is an intellectual disability and endocrine disorder caused by plant homeodomain finger 6 (PHF6) mutations. Individuals with BFLS present with short stature. We report a mouse model of BFLS, in which deletion of Phf6 causes a proportional reduction in body size compared with control mice. Growth hormone (GH) levels were reduced in the absence of PHF6. Phf6-/Y animals displayed a reduction in the expression of the genes encoding GH-releasing hormone (GHRH) in the brain, GH in the pituitary gland and insulin-like growth factor 1 (IGF1) in the liver. Phf6 deletion specifically in the nervous system caused a proportional growth defect, indicating a neuroendocrine contribution to the phenotype. Loss of suppressor of cytokine signaling 2 (SOCS2), a negative regulator of growth hormone signaling partially rescued body size, supporting a reversible deficiency in GH signaling. These results demonstrate that PHF6 regulates the GHRH/GH/IGF1 axis.


Subject(s)
Down-Regulation , Epilepsy/metabolism , Face/abnormalities , Fingers/abnormalities , Growth Disorders/metabolism , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/metabolism , Hypogonadism/metabolism , Insulin-Like Growth Factor I/metabolism , Mental Retardation, X-Linked/metabolism , Obesity/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Animals, Newborn , Disease Models, Animal , Epilepsy/blood , Epilepsy/pathology , Face/pathology , Fingers/pathology , Growth Disorders/blood , Growth Disorders/pathology , Growth Hormone/blood , Hypogonadism/blood , Hypogonadism/pathology , Hypothalamus/metabolism , Insulin-Like Growth Factor I/genetics , Male , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/metabolism , Obesity/blood , Obesity/pathology , Organ Specificity , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
6.
Brain ; 145(12): 4264-4274, 2022 12 19.
Article in English | MEDLINE | ID: mdl-35929549

ABSTRACT

A genetic deficiency of the solute carrier monocarboxylate transporter 8 (MCT8), termed Allan-Herndon-Dudley syndrome, is an important cause of X-linked intellectual and motor disability. MCT8 transports thyroid hormones across cell membranes. While thyroid hormone analogues improve peripheral changes of MCT8 deficiency, no treatment of the neurological symptoms is available so far. Therefore, we tested a gene replacement therapy in Mct8- and Oatp1c1-deficient mice as a well-established model of the disease. Here, we report that targeting brain endothelial cells for Mct8 expression by intravenously injecting the vector AAV-BR1-Mct8 increased tri-iodothyronine (T3) levels in the brain and ameliorated morphological and functional parameters associated with the disease. Importantly, the therapy resulted in a long-lasting improvement in motor coordination. Thus, the data support the concept that MCT8 mediates the transport of thyroid hormones into the brain and indicate that a readily accessible vascular target can help overcome the consequences of the severe disability associated with MCT8 deficiency.


Subject(s)
Disabled Persons , Mental Retardation, X-Linked , Motor Disorders , Symporters , Mice , Animals , Humans , Blood-Brain Barrier/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Muscle Hypotonia/genetics , Muscular Atrophy , Endothelial Cells/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Thyroid Hormones/metabolism , Genetic Therapy , Symporters/genetics , Symporters/metabolism
7.
Int J Mol Sci ; 23(24)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36555189

ABSTRACT

Dysfunctions of the thyroid hormone (TH) transporting monocarboxylate transporter MCT8 lead to a complex X-linked syndrome with abnormal serum TH concentrations and prominent neuropsychiatric symptoms (Allan-Herndon-Dudley syndrome, AHDS). The key features of AHDS are replicated in double knockout mice lacking MCT8 and organic anion transporting protein OATP1C1 (Mct8/Oatp1c1 DKO). In this study, we characterize impairments of brain structure and function in Mct8/Oatp1c1 DKO mice using multimodal magnetic resonance imaging (MRI) and assess the potential of the TH analogue 3,3',5-triiodothyroacetic acid (TRIAC) to rescue this phenotype. Structural and functional MRI were performed in 11-weeks-old male Mct8/Oatp1c1 DKO mice (N = 10), wild type controls (N = 7) and Mct8/Oatp1c1 DKO mice (N = 13) that were injected with TRIAC (400 ng/g bw s.c.) daily during the first three postnatal weeks. Grey and white matter volume were broadly reduced in Mct8/Oatp1c1 DKO mice. TRIAC treatment could significantly improve white matter thinning but did not affect grey matter loss. Network-based statistic showed a wide-spread increase of functional connectivity, while graph analysis revealed an impairment of small-worldness and whole-brain segregation in Mct8/Oatp1c1 DKO mice. Both functional deficits could be substantially ameliorated by TRIAC treatment. Our study demonstrates prominent structural and functional brain alterations in Mct8/Oatp1c1 DKO mice that may underlie the psychomotor deficiencies in AHDS. Additionally, we provide preclinical evidence that early-life TRIAC treatment improves white matter loss and brain network dysfunctions associated with TH transporter deficiency.


Subject(s)
Mental Retardation, X-Linked , Symporters , White Matter , Animals , Male , Mice , White Matter/metabolism , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Thyroid Hormones/metabolism , Muscular Atrophy/metabolism , Mice, Knockout , Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Symporters/genetics , Symporters/metabolism
8.
J Biol Chem ; 295(10): 3247-3256, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31996374

ABSTRACT

Snyder-Robinson syndrome (SRS) is an X-linked intellectual disability syndrome caused by a loss-of-function mutation in the spermine synthase (SMS) gene. Primarily affecting males, the main manifestations of SRS include osteoporosis, hypotonic stature, seizures, cognitive impairment, and developmental delay. Because there is no cure for SRS, treatment plans focus on alleviating symptoms rather than targeting the underlying causes. Biochemically, the cells of individuals with SRS accumulate excess spermidine, whereas spermine levels are reduced. We recently demonstrated that SRS patient-derived lymphoblastoid cells are capable of transporting exogenous spermine and its analogs into the cell and, in response, decreasing excess spermidine pools to normal levels. However, dietary supplementation of spermine does not appear to benefit SRS patients or mouse models. Here, we investigated the potential use of a metabolically stable spermine mimetic, (R,R)-1,12-dimethylspermine (Me2SPM), to reduce the intracellular spermidine pools of SRS patient-derived cells. Me2SPM can functionally substitute for the native polyamines in supporting cell growth while stimulating polyamine homeostatic control mechanisms. We found that both lymphoblasts and fibroblasts from SRS patients can accumulate Me2SPM, resulting in significantly decreased spermidine levels with no adverse effects on growth. Me2SPM administration to mice revealed that Me2SPM significantly decreases spermidine levels in multiple tissues. Importantly, Me2SPM was detectable in brain tissue, the organ most affected in SRS, and was associated with changes in polyamine metabolic enzymes. These findings indicate that the (R,R)-diastereomer of 1,12-Me2SPM represents a promising lead compound in developing a treatment aimed at targeting the molecular mechanisms underlying SRS pathology.


Subject(s)
Mental Retardation, X-Linked/pathology , Spermidine/metabolism , Spermine/analogs & derivatives , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Brain/metabolism , Chromatography, High Pressure Liquid , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Mental Retardation, X-Linked/metabolism , Mice , Mice, Inbred C57BL , Polyamines/analysis , Polyamines/metabolism , Spermine/administration & dosage , Spermine/metabolism , Spermine/pharmacology , Spermine Synthase/genetics , Tumor Cells, Cultured
9.
J Neurochem ; 159(1): 29-60, 2021 10.
Article in English | MEDLINE | ID: mdl-33638179

ABSTRACT

MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections, and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and zcomplex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum, and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.


Subject(s)
Brain/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Animals , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mental Retardation, X-Linked/pathology , Mutation/physiology
10.
Biochemistry ; 59(13): 1367-1377, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32207963

ABSTRACT

More than 80 loss-of-function (LOF) mutations in the SLC6A8 creatine transporter (hCRT1) are responsible for cerebral creatine deficiency syndrome (CCDS), which gives rise to a spectrum of neurological defects, including intellectual disability, epilepsy, and autism spectrum disorder. To gain insight into the nature of the molecular defects caused by these mutations, we quantitatively profiled the cellular processing, trafficking, expression, and function of eight pathogenic CCDS variants in relation to the wild type (WT) and one neutral isoform. All eight CCDS variants exhibit measurable proteostatic deficiencies that likely contribute to the observed LOF. However, the magnitudes of their specific effects on the expression and trafficking of hCRT1 vary considerably, and we find that the LOF associated with two of these variants primarily arises from the disruption of the substrate-binding pocket. In conjunction with an analysis of structural models of the transporter, we use these data to suggest mechanistic classifications for these variants. To evaluate potential avenues for therapeutic intervention, we assessed the sensitivity of these variants to temperature and measured their response to the proteostasis regulator 4-phenylbutyrate (4-PBA). Only one of the tested variants (G132V) is sensitive to temperature, though its response to 4-PBA is negligible. Nevertheless, 4-PBA significantly enhances the activity of WT hCRT1 in HEK293T cells, which suggests it may be worth evaluating as a therapeutic for female intellectual disability patients carrying a single CCDS mutation. Together, these findings reveal that pathogenic SLC6A8 mutations cause a spectrum of molecular defects that should be taken into consideration in future efforts to develop CCDS therapeutics.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Creatine/deficiency , Mental Retardation, X-Linked/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Brain Diseases, Metabolic, Inborn/genetics , Creatine/genetics , Creatine/metabolism , HEK293 Cells , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mental Retardation, X-Linked/genetics , Mutation, Missense , Nerve Tissue Proteins/chemistry , Phenylbutyrates/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism
11.
Mol Psychiatry ; 24(11): 1748-1768, 2019 11.
Article in English | MEDLINE | ID: mdl-29728705

ABSTRACT

RLIM, also known as RNF12, is an X-linked E3 ubiquitin ligase acting as a negative regulator of LIM-domain containing transcription factors and participates in X-chromosome inactivation (XCI) in mice. We report the genetic and clinical findings of 84 individuals from nine unrelated families, eight of whom who have pathogenic variants in RLIM (RING finger LIM domain-interacting protein). A total of 40 affected males have X-linked intellectual disability (XLID) and variable behavioral anomalies with or without congenital malformations. In contrast, 44 heterozygous female carriers have normal cognition and behavior, but eight showed mild physical features. All RLIM variants identified are missense changes co-segregating with the phenotype and predicted to affect protein function. Eight of the nine altered amino acids are conserved and lie either within a domain essential for binding interacting proteins or in the C-terminal RING finger catalytic domain. In vitro experiments revealed that these amino acid changes in the RLIM RING finger impaired RLIM ubiquitin ligase activity. In vivo experiments in rlim mutant zebrafish showed that wild type RLIM rescued the zebrafish rlim phenotype, whereas the patient-specific missense RLIM variants failed to rescue the phenotype and thus represent likely severe loss-of-function mutations. In summary, we identified a spectrum of RLIM missense variants causing syndromic XLID and affecting the ubiquitin ligase activity of RLIM, suggesting that enzymatic activity of RLIM is required for normal development, cognition and behavior.


Subject(s)
Mental Retardation, X-Linked/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adolescent , Adult , Animals , Child , Child, Preschool , Conduct Disorder/genetics , Female , Genes, X-Linked , HEK293 Cells , Humans , Infant, Newborn , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mental Retardation, X-Linked/metabolism , Mice , Middle Aged , Mutation , Pedigree , Transcription Factors/genetics , Ubiquitination , X Chromosome Inactivation , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198194

ABSTRACT

The Thyroid hormone Receptor Interacting Protein 12 (TRIP12) protein belongs to the 28-member Homologous to the E6-AP C-Terminus (HECT) E3 ubiquitin ligase family. First described as an interactor of the thyroid hormone receptor, TRIP12's biological importance was revealed by the embryonic lethality of a murine model bearing an inactivating mutation in the TRIP12 gene. Further studies showed the participation of TRIP12 in the regulation of major biological processes such as cell cycle progression, DNA damage repair, chromatin remodeling, and cell differentiation by an ubiquitination-mediated degradation of key protein substrates. Moreover, alterations of TRIP12 expression have been reported in cancers that can serve as predictive markers of therapeutic response. The TRIP12 gene is also referenced as a causative gene associated to intellectual disorders such as Clark-Baraitser syndrome and is clearly implicated in Autism Spectrum Disorder. The aim of the review is to provide an exhaustive and integrated overview of the different aspects of TRIP12 ranging from its regulation, molecular functions and physio-pathological implications.


Subject(s)
Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Facies , Growth Disorders/genetics , Growth Disorders/metabolism , Humans , Hydrocephalus/genetics , Hydrocephalus/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mutation/genetics , Neoplasms/genetics , Neoplasms/metabolism , Obesity/genetics , Obesity/metabolism
13.
Hum Mol Genet ; 26(23): 4699-4714, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973667

ABSTRACT

Intellectual Disability is a common and heterogeneous disorder characterized by limitations in intellectual functioning and adaptive behaviour, whose molecular mechanisms remain largely unknown. Among the numerous genes found to be involved in the pathogenesis of intellectual disability, 10% are located on the X-chromosome. We identified a missense mutation (c.236 C > G; p.S79W) in the SYN1 gene coding for synapsin I in the MRX50 family, affected by non-syndromic X-linked intellectual disability. Synapsin I is a neuronal phosphoprotein involved in the regulation of neurotransmitter release and neuronal development. Several mutations in SYN1 have been identified in patients affected by epilepsy and/or autism. The S79W mutation segregates with the disease in the MRX50 family and all affected members display intellectual disability as sole clinical manifestation. At the protein level, the S79W Synapsin I mutation is located in the region of the B-domain involved in recognition of highly curved membranes. Expression of human S79W Synapsin I in Syn1 knockout hippocampal neurons causes aberrant accumulation of small clear vesicles in the soma, increased clustering of synaptic vesicles at presynaptic terminals and increased frequency of excitatory spontaneous release events. In addition, the presence of S79W Synapsin I strongly reduces the mobility of synaptic vesicles, with possible implications for the regulation of neurotransmitter release and synaptic plasticity. These results implicate SYN1 in the pathogenesis of non-syndromic intellectual disability, showing that alterations of synaptic vesicle trafficking are one possible cause of this disease, and suggest that distinct mutations in SYN1 may lead to distinct brain pathologies.


Subject(s)
Mental Retardation, X-Linked/genetics , Mutation, Missense , Synapsins/genetics , Synaptic Vesicles/genetics , Animals , Base Sequence , Humans , Mental Retardation, X-Linked/metabolism , Mice , Mice, Knockout , Mutation , Neurogenesis/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Pedigree , Presynaptic Terminals/metabolism , Primary Cell Culture , Protein Transport , Synapsins/metabolism , Synaptic Transmission/genetics , Synaptic Vesicles/metabolism
14.
Hum Mol Genet ; 26(5): 955-968, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28073926

ABSTRACT

Renpenning syndrome is a group of X-linked intellectual disability syndromes caused by mutations in human polyglutamine-binding protein 1 (PQBP1) gene. Little is known about the molecular pathogenesis of the various mutations that cause the notable variability in patients. In this study, we examine the cellular and synaptic functions of the most common mutations found in the patients: c.461_462delAG, c.459_462delAGAG and c.463_464dupAG in an AG hexamer in PQBP1 exon 4. We discovered that PQBP1 c.459_462delAGAG and c.463_464dupAG mutations encode a new C-terminal epitope that preferentially binds non-phosphorylated fragile X mental retardation protein (FMRP) and promotes its ubiquitin-mediated degradation. Impairment of FMRP function up-regulates its targets such as MAP1B, and disrupts FMRP-dependent synaptic scaling in primary cultured neurons. In Drosophila neuromuscular junction model, PQBP1 c.463_464dupAG transgenic flies showed remarkable defects of synaptic over-growth, which can be rescued by exogenously expressing dFMRP. Our data strongly support a gain-of-function pathogenic mechanism of PQBP1 c.459_462delAGAG and c.463_464dupAG mutations, and suggest that therapeutic strategies to restore FMRP function may be beneficial for those patients.


Subject(s)
Carrier Proteins/genetics , Cerebral Palsy/genetics , Fragile X Mental Retardation Protein/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Nuclear Proteins/genetics , Animals , Animals, Genetically Modified , Carrier Proteins/biosynthesis , Cerebral Palsy/metabolism , Cerebral Palsy/pathology , DNA-Binding Proteins , Disease Models, Animal , Drosophila/genetics , Epitopes/genetics , Epitopes/immunology , Fragile X Mental Retardation Protein/biosynthesis , Humans , Intellectual Disability/immunology , Intellectual Disability/pathology , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Microtubule-Associated Proteins/genetics , Mutation , Neuromuscular Junction , Nuclear Proteins/biosynthesis , Peptides/genetics , Proteolysis , Ubiquitin/genetics
15.
Endocr J ; 66(1): 19-29, 2019 Jan 28.
Article in English | MEDLINE | ID: mdl-30369548

ABSTRACT

Monocarboxylate transporter 8 (MCT8) facilitates T3 uptake into cells. Mutations in MCT8 lead to Allan-Herndon-Dudley syndrome (AHDS), which is characterized by severe psychomotor retardation and abnormal thyroid hormone profile. Nine uncharacterized MCT8 mutations in Japanese patients with severe neurocognitive impairment and elevated serum T3 levels were studied regarding the transport of T3. Human MCT8 (hMCT8) function was studied in wild-type (WT) or mutant hMCT8-transfected human placental choriocarcinoma cells (JEG3) by visualizing the locations of the proteins in the cells, detecting specific proteins, and measuring T3 uptake. We identified 6 missense (p.Arg445Ser, p.Asp498Asn, p.Gly276Arg, p.Gly196Glu, p.Gly401Arg, and p.Gly312Arg), 2 frameshift (p.Arg355Profs*64 and p.Tyr550Serfs*17), and 1 deletion (p.Pro561del) mutation(s) in the hMCT8 gene. All patients exhibited clinical characteristics of AHDS with high free T3, low-normal free T4, and normal-elevated TSH levels. All tested mutants were expressed at the protein level, except p.Arg355Profs*64 and p.Tyr550Serfs*17, which were truncated, and were inactive in T3 uptake, excluding p.Arg445Ser and p.Pro561del mutants, compared with WT-hMCT8. Immunocytochemistry revealed plasma membrane localization of p.Arg445Ser and p.Pro561del mutants similar with WT-hMCT8. The other mutants failed to localize in significant amount(s) in the plasma membrane and instead localized in the cytoplasm. These data indicate that p.Arg445Ser and p.Pro561del mutants preserve residual function, whereas p.Asp498Asn, p.Gly276Arg, p.Gly196Glu, p.Gly401Arg, p.Gly312Arg, p.Arg355Profs*64, and p.Tyr550Serfs*17 mutants lack function. These findings suggest that the mutations in MCT8 cause loss of function by reducing protein expression, impairing trafficking of protein to plasma membrane, and disrupting substrate channel.


Subject(s)
Cell Membrane/metabolism , Cytoplasm/metabolism , Mental Retardation, X-Linked/genetics , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/genetics , Muscular Atrophy/genetics , Protein Transport/genetics , Triiodothyronine/metabolism , Adolescent , Asian People , Cell Line, Tumor , Child , Child, Preschool , Genetic Vectors , Humans , Immunohistochemistry , In Vitro Techniques , Infant , Japan , Loss of Function Mutation , Male , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/physiopathology , Monocarboxylic Acid Transporters/metabolism , Muscle Hypotonia/metabolism , Muscle Hypotonia/physiopathology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Mutation , Symporters , Thyrotropin/metabolism , Thyroxine/metabolism , Transfection , Young Adult
16.
Hum Mol Genet ; 25(21): 4787-4803, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28173139

ABSTRACT

ATRX is a chromatin remodeling protein that is mutated in several intellectual disability disorders including alpha-thalassemia/mental retardation, X-linked (ATR-X) syndrome. We previously reported the prevalence of ophthalmological defects in ATR-X syndrome patients, and accordingly we find morphological and functional visual abnormalities in a mouse model harboring a mutation occurring in ATR-X patients. The visual system abnormalities observed in these mice parallels the Atrx-null retinal phenotype characterized by interneuron defects and selective loss of amacrine and horizontal cells. The mechanisms that underlie selective neuronal vulnerability and neurodegeneration in the central nervous system upon Atrx mutation or deletion are unknown. To interrogate the cellular specificity of Atrx for its retinal neuroprotective functions, we employed a combination of temporal and lineage-restricted conditional ablation strategies to generate five different conditional knockout mouse models, and subsequently identified a non-cell-autonomous requirement for Atrx in bipolar cells for inhibitory interneuron survival in the retina. Atrx-deficient retinal bipolar cells exhibit functional, structural and molecular alterations consistent with impairments in neuronal activity and connectivity. Gene expression changes in the Atrx-null retina indicate defective synaptic structure and neuronal circuitry, suggest excitotoxic mechanisms of neurodegeneration, and demonstrate that common targets of ATRX in the forebrain and retina may contribute to similar neuropathological processes underlying cognitive impairment and visual dysfunction in ATR-X syndrome.


Subject(s)
Mental Retardation, X-Linked/genetics , X-linked Nuclear Protein/genetics , alpha-Thalassemia/genetics , Animals , Chromatin , Disease Models, Animal , Interneurons/metabolism , Male , Mental Retardation, X-Linked/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Neurons/metabolism , Nuclear Proteins/genetics , Retina/metabolism , Retinal Bipolar Cells/metabolism , X-linked Nuclear Protein/metabolism , alpha-Thalassemia/metabolism
17.
Mol Genet Metab ; 123(4): 463-471, 2018 04.
Article in English | MEDLINE | ID: mdl-29478817

ABSTRACT

Creatine transporter is currently the focus of renewed interest with emerging roles in brain neurotransmission and physiology, and the bioenergetics of cancer metastases. We here report on amendments of a standard creatine uptake assay which might help clinical chemistry laboratories to extend their current range of measurements of creatine and metabolites in body fluids to functional enzyme explorations. In this respect, short incubation times and the use of a stable-isotope-labeled substrate (D3-creatine) preceded by a creatine wash-out step from cultured fibroblast cells by removal of fetal bovine serum (rich in creatine) from the incubation medium are recommended. Together, these measures decreased, by a first order of magnitude, creatine concentrations in the incubation medium at the start of creatine-uptake studies and allowed to functionally discriminate between 4 hemizygous male and 4 heterozygous female patients with X-linked SLC6A8 deficiency, and between this cohort of eight patients and controls. The functional assay corroborated genetic diagnosis of SLC6A8 deficiency. Gene anomalies in our small cohort included splicing site (c.912G > A [p.Ile260_Gln304del], c.778-2A > G and c.1495 + 2 T > G), substitution (c.407C > T) [p.Ala136Val] and deletion (c.635_636delAG [p.Glu212Valfs*84] and c.1324delC [p.Gln442Lysfs*21]) variants with reduced creatine transporter function validating their pathogenicity, including that of a previously unreported c.1324delC variant. The present assay adaptations provide an easy, reliable and discriminative manner for exploring creatine transporter activity and disease variations. It might apply to drug testing or other evaluations in the genetic and metabolic horizons covered by the emerging functions of creatine and its transporter, in a way, however, requiring and completed by additional studies on female patients and blood-brain barrier permeability properties of selected compounds. As a whole, the proposed assay of creatine transporter positively adds to currently existing measurements of this transporter activity, and determining on a large scale the extent of its exact suitability to detect female patients should condition in the future its transfer in clinical practice.


Subject(s)
Brain Diseases, Metabolic, Inborn/metabolism , Creatine/deficiency , Fibroblasts/metabolism , Mental Retardation, X-Linked/metabolism , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Adolescent , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/pathology , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Creatine/genetics , Creatine/metabolism , Female , Fibroblasts/pathology , Follow-Up Studies , Humans , Infant , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Prognosis
18.
Gen Comp Endocrinol ; 265: 219-229, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29183795

ABSTRACT

Monocarboxylate transporter 8 (MCT8) facilitates transmembrane transport of thyroid hormones (THs) ensuring their action on gene expression during vertebrate neurodevelopment. A loss of MCT8 in humans results in severe psychomotor deficits associated with the Allan-Herndon-Dudley Syndrome (AHDS). However, where and when exactly a lack of MCT8 causes the neurological manifestations remains unclear because of the varying expression pattern of MCT8 between specific brain regions and cells. Here, we elaborate on the animal models that have been generated to elucidate the mechanisms underlying MCT8-deficient brain development. The absence of a clear neurological phenotype in Mct8 knockout mice made it clear that a single species would not suffice. The evolutionary conservation of TH action on neurodevelopment as well as the components regulating TH signalling however offers the opportunity to answer different aspects of MCT8 function in brain development using different vertebrate species. Moreover, the plethora of tools for genome editing available today facilitates gene silencing in these animals as well. Studies in the recently generated mct8-deficient zebrafish and Mct8/Oatp1c1 double knockout mice have put forward the current paradigm of impaired TH uptake at the level of the blood-brain barrier during peri- and postnatal development as being the main pathophysiological mechanism of AHDS. RNAi vector-based, cell-specific induction of MCT8 knockdown in the chicken embryo points to an additional function of MCT8 at the level of the neural progenitors during early brain development. Future studies including also additional in vivo models like Xenopus or in vitro approaches such as induced pluripotent stem cells will continue to help unravelling the exact role of MCT8 in developmental events. In the end, this multispecies approach will lead to a unifying thesis regarding the cellular and molecular mechanisms responsible for the neurological phenotype in AHDS patients.


Subject(s)
Brain/embryology , Brain/metabolism , Monocarboxylic Acid Transporters/metabolism , Thyroid Hormones/metabolism , Zebrafish/metabolism , Animals , Humans , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Models, Biological , Muscle Hypotonia/metabolism , Muscle Hypotonia/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology
19.
Neurobiol Dis ; 108: 298-306, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28887151

ABSTRACT

Following exocytosis, synaptic vesicles (SVs) have to be reformed with the correct complement of proteins in the correct stoichiometry to ensure continued neurotransmission. Synaptophysin is a highly abundant, integral SV protein necessary for the efficient retrieval of the SV SNARE protein, synaptobrevin II (sybII). However the molecular mechanism underpinning synaptophysin-dependent sybII retrieval is still unclear. We recently identified a male patient with severe intellectual disability, hypotonia, epilepsy and callosal agenesis who has a point mutation in the juxtamembrane region of the fourth transmembrane domain of synaptophysin (T198I). This mutation had no effect on the activity-dependent retrieval of synaptophysin that was tagged with the genetically-encoded pH-sensitive reporter (pHluorin) in synaptophysin knockout hippocampal cultures. This suggested the mutant has no global effect on SV endocytosis, which was confirmed when retrieval of a different SV cargo (the glutamate transporter vGLUT1) was examined. However neurons expressing this T198I mutant did display impaired activity-dependent sybII retrieval, similar to that observed in synaptophysin knockout neurons. Interestingly this impairment did not result in an increased stranding of sybII at the plasma membrane. Screening of known human synaptophysin mutations revealed a similar presynaptic phenotype between T198I and a mutation found in X-linked intellectual disability. Thus this novel human synaptophysin mutation has revealed that aberrant retrieval and increased plasma membrane localisation of SV cargo can be decoupled in human disease.


Subject(s)
Biological Transport, Active/physiology , Developmental Disabilities/metabolism , Neurons/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/pathology , Cells, Cultured , Child , Developmental Disabilities/genetics , Endocytosis/physiology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurons/pathology , Synaptic Vesicles/metabolism , Synaptic Vesicles/pathology , Synaptophysin/deficiency , Synaptophysin/genetics
20.
Hum Mol Genet ; 24(7): 1824-35, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25452430

ABSTRACT

ATRX is a chromatin remodeling protein involved in deposition of the histone variant H3.3 at telomeres and pericentromeric heterochromatin. It also influences the expression level of specific genes; however, deposition of H3.3 at transcribed genes is currently thought to occur independently of ATRX. We focused on a set of genes, including the autism susceptibility gene Neuroligin 4 (Nlgn4), that exhibit decreased expression in ATRX-null cells to investigate the mechanisms used by ATRX to promote gene transcription. Overall TERRA levels, as well as DNA methylation and histone modifications at ATRX target genes are not altered and thus cannot explain transcriptional dysregulation. We found that ATRX does not associate with the promoter of these genes, but rather binds within regions of the gene body corresponding to high H3.3 occupancy. These intragenic regions consist of guanine-rich DNA sequences predicted to form non-B DNA structures called G-quadruplexes during transcriptional elongation. We demonstrate that ATRX deficiency corresponds to reduced H3.3 incorporation and stalling of RNA polymerase II at these G-rich intragenic sites. These findings suggest that ATRX promotes the incorporation of histone H3.3 at particular transcribed genes and facilitates transcriptional elongation through G-rich sequences. The inability to transcribe genes such as Nlgn4 could cause deficits in neuronal connectivity and cognition associated with ATRX mutations in humans.


Subject(s)
DNA Helicases/metabolism , Gene Expression Regulation , Guanine/metabolism , Mental Retardation, X-Linked/metabolism , Nuclear Proteins/metabolism , Transcription, Genetic , alpha-Thalassemia/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , DNA Helicases/genetics , DNA Methylation , G-Quadruplexes , Histones/metabolism , Humans , Male , Mental Retardation, X-Linked/embryology , Mental Retardation, X-Linked/genetics , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Open Reading Frames , Promoter Regions, Genetic , X-linked Nuclear Protein , alpha-Thalassemia/embryology , alpha-Thalassemia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL