Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 19(6): e1010800, 2023 06.
Article in English | MEDLINE | ID: mdl-37363915

ABSTRACT

The phosphatase FIG4 and the scaffold protein VAC14 function in the biosynthesis of PI(3,5)P2, a signaling lipid that inhibits the lysosomal chloride transporter ClC-7. Loss-of-function mutations of FIG4 and VAC14 reduce PI(3,5)P2 and result in lysosomal disorders characterized by accumulation of enlarged lysosomes and neurodegeneration. Similarly, a gain of function mutation of CLCN7 encoding ClC-7 also results in enlarged lysosomes. We therefore tested the ability of reduced CLCN7 expression to compensate for loss of FIG4 or VAC14. Knock-out of CLCN7 corrected lysosomal swelling and partially corrected lysosomal hyperacidification in FIG4 null cell cultures. Knockout of the related transporter CLCN6 (ClC-6) in FIG4 null cells did not affect the lysosome phenotype. In the Fig4 null mouse, reduction of ClC-7 by expression of the dominant negative CLCN7 variant p.Gly215Arg improved growth and neurological function and increased lifespan by 20%. These observations demonstrate a role for the CLCN7 chloride transporter in pathogenesis of FIG4 and VAC14 disorders. Reduction of CLCN7 provides a new target for treatment of FIG4 and VAC14 deficiencies that lack specific therapies, such as Charcot-Marie-Tooth Type 4J and Yunis-Varón syndrome.


Subject(s)
Antiporters , Chlorides , Animals , Mice , Antiporters/metabolism , Chlorides/metabolism , Flavoproteins/genetics , Flavoproteins/metabolism , Lysosomes/metabolism , Mice, Knockout , Phosphoinositide Phosphatases/genetics , Phosphoinositide Phosphatases/metabolism , Phosphoric Monoester Hydrolases/genetics
2.
Genet Med ; 26(5): 101097, 2024 05.
Article in English | MEDLINE | ID: mdl-38334070

ABSTRACT

PURPOSE: Pathogenic variants of FIG4 generate enlarged lysosomes and neurological and developmental disorders. To identify additional genes regulating lysosomal volume, we carried out a genome-wide activation screen to detect suppression of enlarged lysosomes in FIG4-/- cells. METHODS: The CRISPR-a gene activation screen utilized sgRNAs from the promoters of protein-coding genes. Fluorescence-activated cell sorting separated cells with correction of the enlarged lysosomes from uncorrected cells. Patient variants of SLC12A9 were identified by exome or genome sequencing and studied by segregation analysis and clinical characterization. RESULTS: Overexpression of SLC12A9, a solute co-transporter, corrected lysosomal swelling in FIG4-/- cells. SLC12A9 (NP_064631.2) colocalized with LAMP2 at the lysosome membrane. Biallelic variants of SLC12A9 were identified in 3 unrelated probands with neurodevelopmental disorders. Common features included intellectual disability, skeletal and brain structural abnormalities, congenital heart defects, and hypopigmented hair. Patient 1 was homozygous for nonsense variant p.(Arg615∗), patient 2 was compound heterozygous for p.(Ser109Lysfs∗20) and a large deletion, and proband 3 was compound heterozygous for p.(Glu290Glyfs∗36) and p.(Asn552Lys). Fibroblasts from proband 1 contained enlarged lysosomes that were corrected by wild-type SLC12A9 cDNA. Patient variant p.(Asn552Lys) failed to correct the lysosomal defect. CONCLUSION: Impaired function of SLC12A9 results in enlarged lysosomes and a recessive disorder with a recognizable neurodevelopmental phenotype.


Subject(s)
Lysosomes , Neurodevelopmental Disorders , Sodium-Potassium-Chloride Symporters , Child , Child, Preschool , Female , Humans , Infant , Male , Alleles , Loss of Function Mutation/genetics , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Pedigree , Phenotype , Sodium-Potassium-Chloride Symporters/genetics
3.
Mol Genet Metab ; 137(4): 382-387, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36434903

ABSTRACT

Loss-of-function mutations of FIG4 impair the biosynthesis of PI(3,5)P2 and are responsible for rare genetic disorders including Yunis-Varón Syndrome and Charcot-Marie-Tooth Disease Type 4 J. Cultured cells deficient in FIG4 accumulate enlarged lysosomes with hyperacidic pH, due in part to impaired regulation of lysosomal ion channels and elevated intra-lysosomal osmotic pressure. We evaluated the effects of the FDA approved drug chloroquine, which is known to reduce lysosome acidity, on FIG4 deficient cell culture and on a mouse model. Chloroquine corrected the enlarged lysosomes in FIG4 null cells. In null mice, addition of chloroquine to the drinking water slowed progression of the disorder. Growth and mobility were dramatically improved during the first month of life, and spongiform degeneration of the nervous system was reduced. The median survival of Fig4 null mice was increased from 4 weeks for untreated mutants to 8 weeks with chloroquine treatment (p < 0.009). Chloroquine thus corrects the lysosomal swelling in cultured cells and ameliorates Fig4 deficiency in vivo. The improved phenotype of mice with complete loss of Fig4 suggests that chloroquine could be beneficial FIG2 in partial loss-of-function disorders such as Charcot-Marie-Tooth Type 4 J.


Subject(s)
Chloroquine , Cleidocranial Dysplasia , Animals , Mice , Chloroquine/pharmacology , Lymphocytes, Null , Cleidocranial Dysplasia/genetics , Lysosomes , Mice, Knockout , Phosphoinositide Phosphatases/genetics , Flavoproteins/genetics
4.
J Cell Sci ; 132(5)2019 02 28.
Article in English | MEDLINE | ID: mdl-30709920

ABSTRACT

The metabolism of PI(3,5)P2 is regulated by the PIKfyve, VAC14 and FIG4 complex, mutations in which are associated with hypopigmentation in mice. These pigmentation defects indicate a key, but as yet unexplored, physiological relevance of this complex in the biogenesis of melanosomes. Here, we show that PIKfyve activity regulates formation of amyloid matrix composed of PMEL protein within the early endosomes in melanocytes, called stage I melanosomes. PIKfyve activity controls the membrane remodeling of stage I melanosomes, which regulates PMEL abundance, sorting and processing. PIKfyve activity also affects stage I melanosome kiss-and-run interactions with lysosomes, which are required for PMEL amyloidogenesis and the establishment of melanosome identity. Mechanistically, PIKfyve activity promotes both the formation of membrane tubules from stage I melanosomes and their release by modulating endosomal actin branching. Taken together, our data indicate that PIKfyve activity is a key regulator of the melanosomal import-export machinery that fine tunes the formation of functional amyloid fibrils in melanosomes and the maintenance of melanosome identity.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Flavoproteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Melanocytes/metabolism , Melanosomes/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide Phosphatases/metabolism , Retinal Pigment Epithelium/metabolism , Amyloid/metabolism , Animals , Cells, Cultured , Flavoproteins/genetics , Homeostasis , Intracellular Signaling Peptides and Proteins/genetics , Melanocytes/pathology , Melanosomes/ultrastructure , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide Phosphatases/genetics , Protein Transport , Retinal Pigment Epithelium/pathology , gp100 Melanoma Antigen/metabolism
5.
Ann Neurol ; 87(3): 339-346, 2020 03.
Article in English | MEDLINE | ID: mdl-31943325

ABSTRACT

OBJECTIVE: SCN8A encephalopathy is a developmental and epileptic encephalopathy (DEE) caused by de novo gain-of-function mutations of sodium channel Nav 1.6 that result in neuronal hyperactivity. Affected individuals exhibit early onset drug-resistant seizures, developmental delay, and cognitive impairment. This study was carried out to determine whether reducing the abundance of the Scn8a transcript with an antisense oligonucleotide (ASO) would delay seizure onset and prolong survival in a mouse model of SCN8A encephalopathy. METHODS: ASO treatment was tested in a conditional mouse model with Cre-dependent expression of the pathogenic patient SCN8A mutation p.Arg1872Trp (R1872W). This model exhibits early onset of seizures, rapid progression, and 100% penetrance. An Scn1a +/- haploinsufficient mouse model of Dravet syndrome was also treated. ASO was administered by intracerebroventricular injection at postnatal day 2, followed in some cases by stereotactic injection at postnatal day 30. RESULTS: We observed a dose-dependent increase in length of survival from 15 to 65 days in the Scn8a-R1872W/+ mice treated with ASO. Electroencephalographic recordings were normal prior to seizure onset. Weight gain and activity in an open field were unaffected, but treated mice were less active in a wheel running assay. A single treatment with Scn8a ASO extended survival of Dravet syndrome mice from 3 weeks to >5 months. INTERPRETATION: Reduction of Scn8a transcript by 25 to 50% delayed seizure onset and lethality in mouse models of SCN8A encephalopathy and Dravet syndrome. Reduction of SCN8A transcript is a promising approach to treatment of intractable childhood epilepsies. Ann Neurol 2020;87:339-346.


Subject(s)
Brain Diseases/prevention & control , Epilepsies, Myoclonic/prevention & control , NAV1.6 Voltage-Gated Sodium Channel/drug effects , Animals , Brain Diseases/complications , Brain Diseases/mortality , Dose-Response Relationship, Drug , Epilepsies, Myoclonic/complications , Epilepsies, Myoclonic/mortality , Female , Infusions, Intraventricular , Male , Mice , Mice, Transgenic , Mutation , NAV1.6 Voltage-Gated Sodium Channel/administration & dosage , Oligonucleotides, Antisense/pharmacology , Seizures/complications , Seizures/prevention & control
6.
Hum Mol Genet ; 27(14): 2443-2453, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29688489

ABSTRACT

The signaling lipid phosphatidylinositol 3,5-bisphosphate, PI(3,5)P2, functions in vesicular trafficking through the endo-lysosomal compartment. Cellular levels of PI(3,5)P2 are regulated by an enzyme complex comprised of the kinase PIKFYVE, the phosphatase FIG4, and the scaffold protein VAC14. Mutations of human FIG4 cause inherited disorders including Charcot-Marie-Tooth disease type 4J, polymicrogyria with epilepsy, and Yunis-Varón syndrome. Constitutive Fig4-/- mice exhibit intention tremor, spongiform degeneration of neural tissue, hypomyelination, and juvenile lethality. To determine whether PI(3,5)P2 is required in the adult, we generated Fig4flox/-; CAG-creER mice and carried out tamoxifen-induced gene ablation. Global ablation in adulthood leads to wasting, tremor, and motor impairment. Death follows within 2 months of tamoxifen treatment, demonstrating a life-long requirement for Fig4. Histological examinations of the sciatic nerve revealed profound Wallerian degeneration of myelinated fibers, but not C-fiber axons in Remak bundles. In optic nerve sections, myelinated fibers appear morphologically intact and carry compound action potentials at normal velocity and amplitude. However, when iKO mice are challenged with a chemical white matter lesion, repair of damaged CNS myelin is significantly delayed, demonstrating a novel role for Fig4 in remyelination. Thus, in the adult PNS Fig4 is required to protect myelinated axons from Wallerian degeneration. In the adult CNS, Fig4 is dispensable for fiber stability and nerve conduction, but is required for the timely repair of damaged white matter. The greater vulnerability of the PNS to Fig4 deficiency in the mouse is consistent with clinical observations in patients with Charcot-Marie-Tooth disease.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Flavoproteins/genetics , Nervous System/metabolism , Phosphoinositide Phosphatases/genetics , Phosphoric Monoester Hydrolases/genetics , Animals , Axons/pathology , Central Nervous System/physiopathology , Charcot-Marie-Tooth Disease/physiopathology , Cleidocranial Dysplasia/genetics , Cleidocranial Dysplasia/physiopathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/physiopathology , Humans , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/physiopathology , Mice , Mice, Transgenic , Micrognathism/genetics , Micrognathism/physiopathology , Mutation , Nervous System/pathology , Neurons/pathology , Peripheral Nervous System/physiopathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Polymicrogyria/genetics , Polymicrogyria/physiopathology , Sciatic Nerve/physiopathology
7.
Hum Mutat ; 40(5): 619-630, 2019 05.
Article in English | MEDLINE | ID: mdl-30740813

ABSTRACT

The lipid phosphatase gene FIG4 is responsible for Yunis-Varón syndrome and Charcot-Marie-Tooth disease Type 4J, a peripheral neuropathy. We now describe four families with FIG4 variants and prominent abnormalities of central nervous system (CNS) white matter (leukoencephalopathy), with onset in early childhood, ranging from severe hypomyelination to mild undermyelination, in addition to peripheral neuropathy. Affected individuals inherited biallelic FIG4 variants from heterozygous parents. Cultured fibroblasts exhibit enlarged vacuoles characteristic of FIG4 dysfunction. Two unrelated families segregate the same G > A variant in the +1 position of intron 21 in the homozygous state in one family and compound heterozygous in the other. This mutation in the splice donor site of exon 21 results in read-through from exon 20 into intron 20 and truncation of the final 115 C-terminal amino acids of FIG4, with retention of partial function. The observed CNS white matter disorder in these families is consistent with the myelination defects in the FIG4 null mouse and the known role of FIG4 in oligodendrocyte maturation. The families described here the expanded clinical spectrum of FIG4 deficiency to include leukoencephalopathy.


Subject(s)
Alleles , Demyelinating Diseases/diagnosis , Demyelinating Diseases/genetics , Flavoproteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Phosphoric Monoester Hydrolases/genetics , Child , Child, Preschool , DNA Mutational Analysis , Demyelinating Diseases/metabolism , Fibroblasts/metabolism , Genotype , Humans , Inheritance Patterns , Magnetic Resonance Imaging , Male , Neuroimaging , Pedigree , Phenotype
8.
Am J Hum Genet ; 99(1): 188-94, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27292112

ABSTRACT

In the PI(3,5)P2 biosynthetic complex, the lipid kinase PIKFYVE and the phosphatase FIG4 are bound to the dimeric scaffold protein VAC14, which is composed of multiple heat-repeat domains. Mutations of FIG4 result in the inherited disorders Charcot-Marie-Tooth disease type 4J, Yunis-Varón syndrome, and polymicrogyria with seizures. We here describe inherited variants of VAC14 in two unrelated children with sudden onset of a progressive neurological disorder and regression of developmental milestones. Both children developed impaired movement with dystonia, became nonambulatory and nonverbal, and exhibited striatal abnormalities on MRI. A diagnosis of Leigh syndrome was rejected due to normal lactate profiles. Exome sequencing identified biallelic variants of VAC14 that were inherited from unaffected heterozygous parents in both families. Proband 1 inherited a splice-site variant that results in skipping of exon 13, p.Ile459Profs(∗)4 (not reported in public databases), and the missense variant p.Trp424Leu (reported in the ExAC database in a single heterozygote). Proband 2 inherited two missense variants in the dimerization domain of VAC14, p.Ala582Ser and p.Ser583Leu, that have not been previously reported. Cultured skin fibroblasts exhibited the accumulation of vacuoles that is characteristic of PI(3,5)P2 deficiency. Vacuolization of fibroblasts was rescued by transfection of wild-type VAC14 cDNA. The similar age of onset and neurological decline in the two unrelated children define a recessive disorder resulting from compound heterozygosity for deleterious variants of VAC14.


Subject(s)
Alleles , Membrane Proteins/genetics , Mutation , Nervous System Diseases/genetics , Age of Onset , Amino Acid Sequence , Child , Child, Preschool , Exome/genetics , Exons/genetics , Female , Genes, Recessive , Heterozygote , Humans , Infant , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/chemistry , Mutation, Missense/genetics , Pedigree
9.
Hum Mol Genet ; 25(2): 340-7, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26604144

ABSTRACT

The lipid phosphatase FIG4 is a subunit of the protein complex that regulates biosynthesis of the signaling lipid PI(3,5)P2. Mutations of FIG4 result in juvenile lethality and spongiform neurodegeneration in the mouse, and are responsible for the human disorders Charcot-Marie-Tooth disease, Yunis-Varon syndrome and polymicrogyria with seizures. We previously demonstrated that conditional expression of a wild-type FIG4 transgene in neurons is sufficient to rescue most of the abnormalities of Fig4 null mice, including juvenile lethality and extensive neurodegeneration. To evaluate the contribution of the phosphatase activity to the in vivo function of Fig4, we introduced the mutation p.Cys486Ser into the Sac phosphatase active-site motif CX5RT. Transfection of the Fig4(Cys486Ser) cDNA into cultured Fig4(-/-) fibroblasts was effective in preventing vacuolization. The neuronal expression of an NSE-Fig4(Cys486Ser) transgene in vivo prevented the neonatal neurodegeneration and juvenile lethality seen in Fig4 null mice. These observations demonstrate that the catalytically inactive FIG4 protein provides significant function, possibly by stabilization of the PI(3,5)P2 biosynthetic complex and/or localization of the complex to endolysosomal vesicles. Despite this partial rescue, later in life the NSE-Fig4(Cys486Ser) transgenic mice display significant abnormalities that include hydrocephalus, defective myelination and reduced lifespan. The late onset phenotype of the NSE-Fig4(Cys486Ser) transgenic mice demonstrates that the phosphatase activity of FIG4 has an essential role in vivo.


Subject(s)
Flavoproteins/genetics , Hydrocephalus/genetics , Mutation , Neurons/metabolism , Animals , Catalytic Domain/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Cleidocranial Dysplasia/genetics , Cleidocranial Dysplasia/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Flavoproteins/metabolism , Hydrocephalus/metabolism , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Mice , Mice, Transgenic , Micrognathism/genetics , Micrognathism/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoinositide Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Polymicrogyria/genetics , Polymicrogyria/metabolism , Schwann Cells/metabolism
10.
Hum Mol Genet ; 24(2): 383-96, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25187576

ABSTRACT

Mutations of FIG4 are responsible for Yunis-Varón syndrome, familial epilepsy with polymicrogyria, and Charcot-Marie-Tooth type 4J neuropathy (CMT4J). Although loss of the FIG4 phospholipid phosphatase consistently causes decreased PtdIns(3,5)P2 levels, cell-specific sensitivity to partial loss of FIG4 function may differentiate FIG4-associated disorders. CMT4J is an autosomal recessive neuropathy characterized by severe demyelination and axonal loss in human, with both motor and sensory involvement. However, it is unclear whether FIG4 has cell autonomous roles in both motor neurons and Schwann cells, and how loss of FIG4/PtdIns(3,5)P2-mediated functions contribute to the pathogenesis of CMT4J. Here, we report that mice with conditional inactivation of Fig4 in motor neurons display neuronal and axonal degeneration. In contrast, conditional inactivation of Fig4 in Schwann cells causes demyelination and defects in autophagy-mediated degradation. Moreover, Fig4-regulated endolysosomal trafficking in Schwann cells is essential for myelin biogenesis during development and for proper regeneration/remyelination after injury. Our data suggest that impaired endolysosomal trafficking in both motor neurons and Schwann cells contributes to CMT4J neuropathy.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Flavoproteins/metabolism , Motor Neurons/metabolism , Schwann Cells/metabolism , Animals , Charcot-Marie-Tooth Disease/genetics , Endosomes/metabolism , Flavoproteins/genetics , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Phosphatidylinositols/metabolism , Phosphoinositide Phosphatases , Protein Transport
11.
Am J Hum Genet ; 92(5): 781-91, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623387

ABSTRACT

Yunis-Varón syndrome (YVS) is an autosomal-recessive disorder with cleidocranial dysplasia, digital anomalies, and severe neurological involvement. Enlarged vacuoles are found in neurons, muscle, and cartilage. By whole-exome sequencing, we identified frameshift and missense mutations of FIG4 in affected individuals from three unrelated families. FIG4 encodes a phosphoinositide phosphatase required for regulation of PI(3,5)P(2) levels, and thus endosomal trafficking and autophagy. In a functional assay, both missense substitutions failed to correct the vacuolar phenotype of Fig4-null mouse fibroblasts. Homozygous Fig4-null mice exhibit features of YVS, including neurodegeneration and enlarged vacuoles in neurons. We demonstrate that Fig4-null mice also have small skeletons with reduced trabecular bone volume and cortical thickness and that cultured osteoblasts accumulate large vacuoles. Our findings demonstrate that homozygosity or compound heterozygosity for null mutations of FIG4 is responsible for YVS, the most severe known human phenotype caused by defective phosphoinositide metabolism. In contrast, in Charcot-Marie-Tooth disease type 4J (also caused by FIG4 mutations), one of the FIG4 alleles is hypomorphic and disease is limited to the peripheral nervous system. This genotype-phenotype correlation demonstrates that absence of FIG4 activity leads to central nervous system dysfunction and extensive skeletal anomalies. Our results describe a role for PI(3,5)P(2) signaling in skeletal development and maintenance.


Subject(s)
Bone Development/genetics , Cleidocranial Dysplasia/genetics , Ectodermal Dysplasia/genetics , Flavoproteins/genetics , Genetic Predisposition to Disease/genetics , Limb Deformities, Congenital/genetics , Micrognathism/genetics , Animals , Base Sequence , Cleidocranial Dysplasia/pathology , Ectodermal Dysplasia/pathology , Exome/genetics , Fibroblasts , Frameshift Mutation/genetics , Genotype , Humans , Limb Deformities, Congenital/pathology , Mice , Micrognathism/pathology , Molecular Sequence Data , Mutation, Missense/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases , Sequence Analysis, DNA
13.
Proc Natl Acad Sci U S A ; 109(43): 17472-7, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23047693

ABSTRACT

Mutations that cause defects in levels of the signaling lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P(2)] lead to profound neurodegeneration in mice. Moreover, mutations in human FIG4 predicted to lower PI(3,5)P(2) levels underlie Charcot-Marie-Tooth type 4J neuropathy and are present in selected cases of amyotrophic lateral sclerosis. In yeast and mammals, PI(3,5)P(2) is generated by a protein complex that includes the lipid kinase Fab1/Pikfyve, the scaffolding protein Vac14, and the lipid phosphatase Fig4. Fibroblasts cultured from Vac14(-/-) and Fig4(-/-) mouse mutants have a 50% reduction in the levels of PI(3,5)P(2), suggesting that there may be PIKfyve-independent pathways that generate this lipid. Here, we characterize a Pikfyve gene-trap mouse (Pikfyve(ß-geo/ß-geo)), a hypomorph with ~10% of the normal level of Pikfyve protein. shRNA silencing of the residual Pikfyve transcript in fibroblasts demonstrated that Pikfyve is required to generate all of the PI(3,5)P(2) pool. Surprisingly, Pikfyve also is responsible for nearly all of the phosphatidylinositol-5-phosphate (PI5P) pool. We show that PI5P is generated directly from PI(3,5)P(2), likely via 3'-phosphatase activity. Analysis of tissues from the Pikfyve(ß-geo/ß-geo) mouse mutants reveals that Pikfyve is critical in neural tissues, heart, lung, kidney, thymus, and spleen. Thus, PI(3,5)P(2) and PI5P have major roles in multiple organs. Understanding the regulation of these lipids may provide insights into therapies for multiple diseases.


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/biosynthesis , Phosphatidylinositol Phosphates/metabolism , Signal Transduction , Animals , Cells, Cultured , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins , Mice , Mice, Mutant Strains , RNA, Messenger/genetics
14.
PLoS Genet ; 7(6): e1002104, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21655088

ABSTRACT

CMT4J is a severe form of Charcot-Marie-Tooth neuropathy caused by mutation of the phosphoinositide phosphatase FIG4/SAC3. Affected individuals are compound heterozygotes carrying the missense allele FIG4-I41T in combination with a null allele. Analysis using the yeast two-hybrid system demonstrated that the I41T mutation impairs interaction of FIG4 with the scaffold protein VAC14. The critical role of this interaction was confirmed by the demonstration of loss of FIG4 protein in VAC14 null mice. We developed a mouse model of CMT4J by expressing a Fig4-I41T cDNA transgene on the Fig4 null background. Expression of the mutant transcript at a level 5 × higher than endogenous Fig4 completely rescued lethality, whereas 2 × expression gave only partial rescue, providing a model of the human disease. The level of FIG4-I41T protein in transgenic tissues is only 2% of that predicted by the transcript level, as a consequence of the protein instability caused by impaired interaction of the mutant protein with VAC14. Analysis of patient fibroblasts demonstrated a comparably low level of mutant I41T protein. The abundance of FIG4-I41T protein in cultured cells is increased by treatment with the proteasome inhibitor MG-132. The data demonstrate that FIG4-I41T is a hypomorphic allele encoding a protein that is unstable in vivo. Expression of FIG4-I41T protein at 10% of normal level is sufficient for long-term survival, suggesting that patients with CMT4J could be treated by increased production or stabilization of the mutant protein. The transgenic model will be useful for testing in vivo interventions to increase the abundance of the mutant protein.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Flavoproteins/genetics , Mutation , Alleles , Animals , Autophagy/genetics , Charcot-Marie-Tooth Disease/metabolism , Fibroblasts/metabolism , Flavoproteins/metabolism , Gliosis/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Mice , Mice, Transgenic , Models, Animal , Phosphoinositide Phosphatases , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Transfection
15.
HGG Adv ; : 100324, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38956874

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed, essential enzymes that complete the first step of protein translation: ligation of amino acids to cognate tRNAs. Genes encoding ARSs have been implicated in myriad dominant and recessive phenotypes, the latter often affecting multiple tissues but with frequent involvement of the central and peripheral nervous systems, liver, and lungs. Threonyl-tRNA synthetase (TARS1) encodes the enzyme that ligates threonine to tRNATHR in the cytoplasm. To date, TARS1 variants have been implicated in a recessive brittle hair phenotype. To better understand TARS1-related recessive phenotypes, we engineered three TARS1 missense variants at conserved residues and studied these variants in S. cerevisiae and C. elegans models. This revealed two loss-of-function variants, including one hypomorphic allele (R433H). We next used R433H to study the effects of partial loss of TARS1 function in a compound heterozygous mouse model (R432H/null). This model presents with phenotypes reminiscent of patients with TARS1 variants and with distinct lung and skin defects. This study expands the potential clinical heterogeneity of TARS1-related recessive disease, which should guide future clinical and genetic evaluations of patient populations.

16.
bioRxiv ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38585737

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed, essential enzymes that complete the first step of protein translation: ligation of amino acids to cognate tRNAs. Genes encoding ARSs have been implicated in myriad dominant and recessive phenotypes, the latter often affecting multiple tissues but with frequent involvement of the central and peripheral nervous system, liver, and lungs. Threonyl-tRNA synthetase (TARS1) encodes the enzyme that ligates threonine to tRNATHR in the cytoplasm. To date, TARS1 variants have been implicated in a recessive brittle hair phenotype. To better understand TARS1-related recessive phenotypes, we engineered three TARS1 missense mutations predicted to cause a loss-of-function effect and studied these variants in yeast and worm models. This revealed two loss-of-function mutations, including one hypomorphic allele (R433H). We next used R433H to study the effects of partial loss of TARS1 function in a compound heterozygous mouse model (R433H/null). This model presents with phenotypes reminiscent of patients with TARS1 variants and with distinct lung and skin defects. This study expands the potential clinical heterogeneity of TARS1-related recessive disease, which should guide future clinical and genetic evaluations of patient populations.

17.
Bipolar Disord ; 15(3): 326-32, 2013 May.
Article in English | MEDLINE | ID: mdl-23551834

ABSTRACT

OBJECTIVE: To investigate the role in bipolar disorder of the C9ORF72 hexanucleotide repeat expansion responsible for frontotemporal lobe dementia and amyotrophic lateral sclerosis. METHODS: Eighty-nine subjects from a previously described panel of individuals with bipolar disorder ascertained for genetic studies were screened to detect expansion of the C9ORF72 repeat. One two-generation family with bipolar disorder and an expanded repeat was characterized in depth using molecular diagnostics, imaging, histopathology, and neurological and neuropsychological evaluation. RESULTS: One proband, with the typical clinical presentation of bipolar disorder, carried an expanded C9ORF72 allele of heterogeneous length between 14 and 20 kilobases (kb) as assessed by Southern blot. The expanded allele was inherited from a parent with atypical, late onset clinical features of bipolar disorder, who subsequently progressed to frontotemporal lobe dementia. The expansion in peripheral blood of the parent ranged from 8.5 to 20 kb. Cultured lymphoblastoid cells from this parent exhibited a homogeneous expansion of only 8.5 kb. CONCLUSIONS: The disease course in the two generations described here demonstrates that expansion of the C9ORF72 may be associated with a form of bipolar disorder that presents clinically with classic phenomenology and progression to neurodegenerative disease. The frequency in our bipolar disorder cohort was only 1%, indicating that C9ORF72 is not a major contributor to bipolar disorder. DNA from cultured cells may be biased towards shorter repeats and nonrepresentative of the endogenous C9ORF72 expansion.


Subject(s)
Bipolar Disorder/genetics , DNA Repeat Expansion/genetics , Family Health , Proteins/genetics , Adult , Bipolar Disorder/complications , Bipolar Disorder/pathology , C9orf72 Protein , Cells, Cultured , Cohort Studies , Disease Progression , Female , Genetic Testing , Humans , Lymphocytes/physiology , Male , Middle Aged , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/genetics , Neuropsychological Tests , Psychiatric Status Rating Scales
18.
G3 (Bethesda) ; 13(8)2023 08 09.
Article in English | MEDLINE | ID: mdl-36691351

ABSTRACT

Loss-of-function mutations of FIG4 are responsible for neurological disorders in human and mouse that result from reduced abundance of the signaling lipid PI(3,5)P2. In contrast, loss-of-function mutations of the phosphoinositide kinase PIP4K2C result in elevated abundance of PI(3,5)P2. These opposing effects on PI(3,5)P2 suggested that we might be able to compensate for deficiency of FIG4 by reducing expression of PIP4K2C. To test this hypothesis in a whole animal model, we generated triallelic mice with genotype Fig 4-/-, Pip4k2c+/-; these mice are null for Fig 4 and haploinsufficient for Pip4k2c. The neonatal lethality of Fig 4 null mice in the C57BL/6J strain background was rescued by reduced expression of Pip4k2c. The lysosome enlargement characteristic of Fig 4 null cells was also reduced by heterozygous loss of Pip4k2c. The data demonstrate interaction between these two genes, and suggest that inhibition of the kinase PIPK4C2 could be a target for treatment of FIG4 deficiency disorders such as Charcot-Marie-Tooth Type 4J and Yunis-Varón Syndrome.


Subject(s)
Cleidocranial Dysplasia , Micrognathism , Mice , Animals , Humans , Mice, Inbred C57BL , Phosphoric Monoester Hydrolases/genetics , Cleidocranial Dysplasia/genetics , Micrognathism/genetics , Phenotype , Phosphatidylinositols , Flavoproteins/genetics , Phosphoinositide Phosphatases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics
19.
J Neurosci ; 31(48): 17736-51, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-22131434

ABSTRACT

The plt (pale tremor) mouse carries a null mutation in the Fig4(Sac3) gene that results in tremor, hypopigmentation, spongiform degeneration of the brain, and juvenile lethality. FIG4 is a ubiquitously expressed phosphatidylinositol 3,5-bisphosphate phosphatase that regulates intracellular vesicle trafficking along the endosomal-lysosomal pathway. In humans, the missense mutation FIG4(I41T) combined with a FIG4 null allele causes Charcot-Marie-Tooth 4J disease, a severe form of peripheral neuropathy. Here we show that Fig4 null mice exhibit a dramatic reduction of myelin in the brain and spinal cord. In the optic nerve, smaller-caliber axons lack myelin sheaths entirely, whereas many large- and intermediate-caliber axons are myelinated but show structural defects at nodes of Ranvier, leading to delayed propagation of action potentials. In the Fig4 null brain and optic nerve, oligodendrocyte (OL) progenitor cells are present at normal abundance and distribution, but the number of myelinating OLs is greatly compromised. The total number of axons in the Fig4 null optic nerve is not reduced. Developmental studies reveal incomplete myelination rather than elevated cell death in the OL linage. Strikingly, there is rescue of CNS myelination and tremor in transgenic mice with neuron-specific expression of Fig4, demonstrating a non-cell-autonomous function of Fig4 in OL maturation and myelin development. In transgenic mice with global overexpression of the human pathogenic FIG4 variant I41T, there is rescue of the myelination defect, suggesting that the CNS of CMT4J patients may be protected from myelin deficiency by expression of the FIG4(I41T) mutant protein.


Subject(s)
Axons/pathology , Charcot-Marie-Tooth Disease/genetics , Flavoproteins/genetics , Myelin Sheath/genetics , Nerve Fibers, Myelinated/pathology , Neurons/metabolism , Phosphoric Monoester Hydrolases/genetics , Animals , Axons/metabolism , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Flavoproteins/metabolism , Mice , Mice, Transgenic , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Fibers, Myelinated/metabolism , Neurons/pathology , Phosphatidylinositols/metabolism , Phosphoinositide Phosphatases , Phosphoric Monoester Hydrolases/metabolism
20.
Am J Hum Genet ; 84(1): 85-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19118816

ABSTRACT

Mutations of the lipid phosphatase FIG4 that regulates PI(3,5)P(2) are responsible for the recessive peripheral-nerve disorder CMT4J. We now describe nonsynonymous variants of FIG4 in 2% (9/473) of patients with amyotrophic lateral sclerosis (ALS) and primary lateral sclerosis (PLS). Heterozygosity for a deleterious allele of FIG4 appears to be a risk factor for ALS and PLS, extending the list of known ALS genes and increasing the clinical spectrum of FIG4-related diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Flavoproteins/genetics , Genetic Predisposition to Disease , Adult , Aged , Amino Acid Sequence , Heterozygote , Humans , Middle Aged , Molecular Sequence Data , Motor Neuron Disease/genetics , Mutation , Phosphoric Monoester Hydrolases
SELECTION OF CITATIONS
SEARCH DETAIL