Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Br J Haematol ; 192(5): 909-921, 2021 03.
Article in English | MEDLINE | ID: mdl-33528045

ABSTRACT

Lowe syndrome (LS) is an oculocerebrorenal syndrome of Lowe (OCRL1) genetic disorder resulting in a defect of the OCRL protein, a phosphatidylinositol-4,5-bisphosphate 5-phosphatase containing various domains including a Rho GTPase-activating protein (RhoGAP) homology domain catalytically inactive. We previously reported surgery-associated bleeding in patients with LS, suggestive of platelet dysfunction, accompanied with a mild thrombocytopenia in several patients. To decipher the role of OCRL in platelet functions and in megakaryocyte (MK) maturation, we conducted a case-control study on 15 patients with LS (NCT01314560). While all had a drastically reduced expression of OCRL, this deficiency did not affect platelet aggregability, but resulted in delayed thrombus formation on collagen under flow conditions, defective platelet spreading on fibrinogen and impaired clot retraction. We evidenced alterations of the myosin light chain phosphorylation (P-MLC), with defective Rac1 activity and, inversely, elevated active RhoA. Altered cytoskeleton dynamics was also observed in cultured patient MKs showing deficient proplatelet extension with increased P-MLC that was confirmed using control MKs transfected with OCRL-specific small interfering(si)RNA (siOCRL). Patients with LS also had an increased proportion of circulating barbell-shaped proplatelets. Our present study establishes that a deficiency of the OCRL protein results in a defective actomyosin cytoskeleton reorganisation in both MKs and platelets, altering both thrombopoiesis and some platelet responses to activation necessary to ensure haemostasis.


Subject(s)
Blood Platelets/cytology , Megakaryocytes/cytology , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/physiology , Thrombopoiesis/physiology , Actomyosin/analysis , Adolescent , Adult , Anemia/etiology , Blood Coagulation , Blood Platelets/ultrastructure , Case-Control Studies , Cell Shape , Child , Collagen , Cytoskeleton/ultrastructure , Female , Gene Silencing , Humans , Male , Megakaryocytes/ultrastructure , Middle Aged , Mutation , Myosin Light Chains/metabolism , Oculocerebrorenal Syndrome/blood , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , Signal Transduction , Thrombocytopenia/etiology , Young Adult
2.
Ann Hum Genet ; 84(6): 456-462, 2020 11.
Article in English | MEDLINE | ID: mdl-32776513

ABSTRACT

BACKGROUND: Variants perturbing the normal splicing of pre-mRNA can lead to human diseases. The splice-altering effect and eventual consequence on gene function was sometimes uncertain and hinders a definitive molecular diagnosis. METHODS: The impact of four rare intronic variants on splicing was analyzed through reverse transcription - polymerase chain reaction (RT-PCR) analysis of mRNA derived from the peripheral blood of patients. The results were compared with in-silico prediction. Potential implication on molecular diagnosis was discussed. RESULTS: Four rare intronic variants of SLC9A6, DLG3, GAA, and OCRL were identified in patients with suspected disorders, respectively. Although these four variants were all predicted to alter splicing by in-silico tools, RT-PCR analysis of mRNA derived from peripheral blood showed these variants affected splicing in different ways: c.899+3_899+6del of SLC9A6 resulted in one-exon skipping and an out-of-frame transcript; c.905-2A > G of DLG3 resulted in a mix of in-frame transcripts; c.1195-11T > A of GAA resulted in the in-frame insertion of nine nucleotides; c.723-2A > C of OCRL resulted in one-exon skipping and in-frame deletion of 102 nucleotides. The consequence revealed by mRNA analysis is essential for accurate interpretation of pathogenicity. CONCLUSION: Four intronic variants all caused aberrant mRNA splicing. For intronic variants with uncertain impact on splicing, mRNA analysis is helpful for ascertainment of alternative splicing and accurate interpretation of pathogenicity.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Mutation , RNA Splicing , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Child, Preschool , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/pathology , Humans , Infant , Male , Microcephaly/genetics , Microcephaly/pathology , Nuclear Proteins/genetics , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , Phenotype , Phosphoric Monoester Hydrolases/genetics , Prognosis , RNA, Messenger/metabolism , Sodium-Hydrogen Exchangers/genetics , Transcription Factors/genetics , alpha-Glucosidases/genetics
3.
Hum Mol Genet ; 29(10): 1700-1715, 2020 06 27.
Article in English | MEDLINE | ID: mdl-32391547

ABSTRACT

Lowe syndrome (LS) is an X-linked developmental disease characterized by cognitive deficiencies, bilateral congenital cataracts and renal dysfunction. Unfortunately, this disease leads to the early death of affected children often due to kidney failure. Although this condition was first described in the early 1950s and the affected gene (OCRL1) was identified in the early 1990s, its pathophysiological mechanism is not fully understood and there is no LS-specific cure available to patients. Here we report two important signaling pathways affected in LS patient cells. While RhoGTPase signaling abnormalities led to adhesion and spreading defects as compared to normal controls, PI3K/mTOR hyperactivation interfered with primary cilia assembly (scenario also observed in other ciliopathies with compromised kidney function). Importantly, we identified two FDA-approved drugs able to ameliorate these phenotypes. Specifically, statins mitigated adhesion and spreading abnormalities while rapamycin facilitated ciliogenesis in LS patient cells. However, no single drug was able to alleviate both phenotypes. Based on these and other observations, we speculate that Ocrl1 has dual, independent functions supporting proper RhoGTPase and PI3K/mTOR signaling. Therefore, this study suggest that Ocrl1-deficiency leads to signaling defects likely to require combinatorial drug treatment to suppress patient phenotypes and symptoms.


Subject(s)
Genetic Diseases, X-Linked/drug therapy , Oculocerebrorenal Syndrome/drug therapy , Phosphoric Monoester Hydrolases/genetics , TOR Serine-Threonine Kinases/genetics , Cell Line , Cilia/drug effects , Cilia/genetics , Cilia/pathology , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , Phenotype , Signal Transduction/drug effects , Sirolimus/pharmacology , rho GTP-Binding Proteins/genetics
4.
Saudi J Kidney Dis Transpl ; 31(1): 285-288, 2020.
Article in English | MEDLINE | ID: mdl-32129227

ABSTRACT

The oculocerebrorenal (OCRL) syndrome, also known as Lowe syndrome (LS), is an X-linked recessive disorder that predominantly affects males and is characterized by growth and mental retardation, congenital cataract and renal Fanconi syndrome. OCRL1 is the gene responsible for LS and encodes an inositol polyphosphate-5-phosphatase. We report a male child from North India, with LS with missense mutation in exon 14 of the OCRL gene.


Subject(s)
Mutation, Missense/genetics , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Adolescent , Brain/diagnostic imaging , Brain/pathology , Hand/diagnostic imaging , Hand/pathology , Humans , India , Male , Oculocerebrorenal Syndrome/diagnostic imaging , Oculocerebrorenal Syndrome/pathology , Pelvis/diagnostic imaging , Pelvis/pathology
5.
Growth Factors ; 37(3-4): 170-177, 2019 08.
Article in English | MEDLINE | ID: mdl-31576764

ABSTRACT

Lowe syndrome is an x-linked disorder characterized by congenital cataracts, nervous system abnormalities and renal tubular dysfunction. With the rising number of reported cases, more patients are found to suffer from endocrine abnormalities. Hereby, three Chinese patients with typical symptoms and extremely short stature were described. The OCRL gene was analyzed. A combination of blood biochemistry and radiological examinations were performed. Growth hormone provocation test was taken in one patient. Nucleotide sequence analysis revealed a de novo novel hemizygous mutation (c.2290_2291delinsCT) in exon 21 in an adolescent boy. As indicated by the growth hormone provocation test, the boy had growth hormone deficiency. The other two patients were brothers with extremely short stature, and manifested the same hemizygous mutation (c.2581G > A) in exon 23. It was speculated that the mutation of OCRL gene could lead to deficiency of growth hormone, for which an early growth hormone intervention may be beneficial.


Subject(s)
Child Development/physiology , Human Growth Hormone/deficiency , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Adolescent , Base Sequence , Body Height/genetics , Child , Child, Preschool , China , Humans , Male , Mutation/genetics , Oculocerebrorenal Syndrome/pathology , Sequence Analysis, DNA
6.
J Cell Biol ; 218(7): 2198-2214, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31118240

ABSTRACT

The tumor suppressor PTEN dephosphorylates PtdIns(3,4,5)P3 into PtdIns(4,5)P2 Here, we make the unexpected discovery that in Drosophila melanogaster PTEN reduces PtdIns(4,5)P2 levels on endosomes, independently of its phosphatase activity. This new PTEN function requires the enzymatic action of dPLCXD, an atypical phospholipase C. Importantly, we discovered that this novel PTEN/dPLCXD pathway can compensate for depletion of dOCRL, a PtdIns(4,5)P2 phosphatase. Mutation of OCRL1, the human orthologue of dOCRL, causes oculocerebrorenal Lowe syndrome, a rare multisystemic genetic disease. Both OCRL1 and dOCRL loss have been shown to promote accumulation of PtdIns(4,5)P2 on endosomes and cytokinesis defects. Here, we show that PTEN or dPLCXD overexpression prevents these defects. In addition, we found that chemical activation of this pathway restores normal cytokinesis in human Lowe syndrome cells and rescues OCRL phenotypes in a zebrafish Lowe syndrome model. Our findings identify a novel PTEN/dPLCXD pathway that controls PtdIns(4,5)P2 levels on endosomes. They also point to a potential new strategy for the treatment of Lowe syndrome.


Subject(s)
Drosophila Proteins/genetics , Oculocerebrorenal Syndrome/genetics , PTEN Phosphohydrolase/genetics , Phosphoric Monoester Hydrolases/genetics , Type C Phospholipases/genetics , Animals , Cytokinesis/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Endosomes/genetics , Endosomes/metabolism , Gene Expression Regulation/genetics , Humans , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Signal Transduction
7.
Cell Mol Life Sci ; 75(21): 4021-4040, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29916093

ABSTRACT

Mitochondrial intracrines are extracellular signaling proteins, targeted to the mitochondria. The pathway for mitochondrial targeting of mitochondrial intracrines and actions in the mitochondria remains unknown. Megalin/LRP2 mediates the uptake of vitamins and proteins, and is critical for clearance of amyloid-ß protein from the brain. Megalin mutations underlie the pathogenesis of Donnai-Barrow and Lowe syndromes, characterized by brain defects and kidney dysfunction; megalin was not previously known to reside in the mitochondria. Here, we show megalin is present in the mitochondria and associates with mitochondrial anti-oxidant proteins SIRT3 and stanniocalcin-1 (STC1). Megalin shuttles extracellularly-applied STC1, angiotensin II and TGF-ß to the mitochondria through the retrograde early endosome-to-Golgi transport pathway and Rab32. Megalin knockout in cultured cells impairs glycolytic and respiratory capacities. Thus, megalin is critical for mitochondrial biology; mitochondrial intracrine signaling is a continuum of the retrograde early endosome-to-Golgi-Rab32 pathway and defects in this pathway may underlie disease processes in many systems.


Subject(s)
Amyloid beta-Peptides/genetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mitochondria/genetics , rab GTP-Binding Proteins/genetics , Agenesis of Corpus Callosum/genetics , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Membrane/genetics , Glycoproteins/genetics , HEK293 Cells , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/metabolism , Hernias, Diaphragmatic, Congenital/pathology , Humans , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mice , Mitochondria/metabolism , Myopia/genetics , Myopia/metabolism , Myopia/pathology , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , RAW 264.7 Cells , Renal Tubular Transport, Inborn Errors/genetics , Renal Tubular Transport, Inborn Errors/metabolism , Renal Tubular Transport, Inborn Errors/pathology , Signal Transduction , Sirtuin 3/genetics , Transforming Growth Factor beta/genetics , rab GTP-Binding Proteins/metabolism
8.
PLoS One ; 13(2): e0192635, 2018.
Article in English | MEDLINE | ID: mdl-29444177

ABSTRACT

Lowe syndrome is an X-linked condition characterized by congenital cataracts, neurological abnormalities and kidney malfunction. This lethal disease is caused by mutations in the OCRL1 gene, which encodes for the phosphatidylinositol 5-phosphatase Ocrl1. While in the past decade we witnessed substantial progress in the identification and characterization of LS patient cellular phenotypes, many of these studies have been performed in knocked-down cell lines or patient's cells from accessible cell types such as skin fibroblasts, and not from the organs affected. This is partially due to the limited accessibility of patient cells from eyes, brain and kidneys. Here we report the preparation of induced pluripotent stem cells (iPSCs) from patient skin fibroblasts and their reprogramming into kidney cells. These reprogrammed kidney cells displayed primary cilia assembly defects similar to those described previously in cell lines. Additionally, the transcription factor and cap mesenchyme marker Six2 was substantially retained in the Golgi complex and the functional nuclear-localized fraction was reduced. These results were confirmed using different batches of differentiated cells from different iPSC colonies and by the use of the human proximal tubule kidney cell line HK2. Indeed, OCRL1 KO led to both ciliogenesis defects and Six2 retention in the Golgi complex. In agreement with Six2's role in the suppression of ductal kidney lineages, cells from this pedigree were over-represented among patient kidney-reprogrammed cells. We speculate that this diminished efficacy to produce cap mesenchyme cells would cause LS patients to have difficulties in replenishing senescent or damaged cells derived from this lineage, particularly proximal tubule cells, leading to pathological scenarios such as tubular atrophy.


Subject(s)
Cell Differentiation , Cilia/pathology , Golgi Apparatus/metabolism , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/pathology , Kidney/pathology , Nerve Tissue Proteins/metabolism , Oculocerebrorenal Syndrome/pathology , Cell Lineage , Humans
9.
Mol Vis ; 24: 847-852, 2018.
Article in English | MEDLINE | ID: mdl-30713423

ABSTRACT

Purpose: To identify the genetic variation in two unrelated probands with congenital cataract and to perform functional analysis of the detected variants. Methods: Clinical examination and phenotyping, segregation, and functional analysis were performed for the two studied pedigrees. Results: A novel OCRL gene variant (c.1964A>T, p. (Asp655Val)) was identified. This variant causes defects in OCRL protein folding and mislocalization to the cytoplasm. In addition, the variant's location close to the Rab binding site is likely to be associated with membrane targeting abnormalities. Conclusions: The results highlight the importance of early genetic diagnosis in infants with congenital cataract and show that mutations in the OCRL gene can present as apparently isolated congenital cataract.


Subject(s)
Cataract/genetics , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Point Mutation , rab GTP-Binding Proteins/genetics , Amino Acid Substitution , Binding Sites , Cataract/congenital , Cataract/metabolism , Cataract/pathology , Child , Gene Expression , Hemizygote , Humans , Male , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Pedigree , Phenotype , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism
10.
PLoS Genet ; 13(10): e1007052, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29028801

ABSTRACT

Lowe Syndrome is a developmental disorder characterized by eye, kidney, and neurological pathologies, and is caused by mutations in the phosphatidylinositol-5-phosphatase OCRL. OCRL plays diverse roles in endocytic and endolysosomal trafficking, cytokinesis, and ciliogenesis, but it is unclear which of these cellular functions underlie specific patient symptoms. Here, we show that mutation of Drosophila OCRL causes cell-autonomous activation of hemocytes, which are macrophage-like cells of the innate immune system. Among many cell biological defects that we identified in docrl mutant hemocytes, we pinpointed the cause of innate immune cell activation to reduced Rab11-dependent recycling traffic and concomitantly increased Rab7-dependent late endosome traffic. Loss of docrl amplifies multiple immune-relevant signals, including Toll, Jun kinase, and STAT, and leads to Rab11-sensitive mis-sorting and excessive secretion of the Toll ligand Spåtzle. Thus, docrl regulation of endosomal traffic maintains hemocytes in a poised, but quiescent state, suggesting mechanisms by which endosomal misregulation of signaling may contribute to symptoms of Lowe syndrome.


Subject(s)
Cytokinesis/genetics , Immunity, Innate/genetics , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Animals , Drosophila , Endosomes/genetics , Endosomes/pathology , Hemocytes/metabolism , Hemocytes/pathology , Humans , Mutation , Oculocerebrorenal Syndrome/pathology , Protein Binding
11.
Sci Rep ; 7(1): 1442, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28473699

ABSTRACT

Mutations in the OCRL1 gene result in the oculocerebrorenal syndrome of Lowe, with symptoms including congenital bilateral cataracts, glaucoma, renal failure, and neurological impairments. OCRL1 encodes an inositol polyphosphate 5-phosphatase which preferentially dephosphorylates phosphatidylinositide 4,5 bisphosphate (PI(4,5)P2). We have identified two novel mutations in two unrelated Lowe syndrome patients with congenital glaucoma. Novel deletion mutations are detected at c.739-742delAAAG in Lowe patient 1 and c.1595-1631del in Lowe patient 2. End stage glaucoma in patient 2 resulted in the enucleation of the eye, which on histology demonstrated corneal keloid, fibrous infiltration of the angle, ectropion uvea, retinal gliosis, and retinal ganglion cell loss. We measured OCRL protein levels in patient keratinocytes and found that Lowe 1 patient cells had significantly reduced OCRL protein as compared to the control keratinocytes. Genotype-phenotype correlation of OCRL1 mutations associated with congenital glaucoma revealed clustering of missense and deletion mutations in the 5-phosphatase domain and the RhoGAP-like domain. In conclusion, we report novel OCRL1 mutations in Lowe syndrome patients and the corresponding histopathologic analysis of one patient's ocular pathology.


Subject(s)
Glaucoma/pathology , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/genetics , Eye/pathology , Genotype , Glaucoma/congenital , Glaucoma/genetics , Humans , Keratinocytes/metabolism , Male , Mutation, Missense , Oculocerebrorenal Syndrome/complications , Pedigree , Phenotype , Phosphoric Monoester Hydrolases/chemistry , Protein Structure, Tertiary , Sequence Deletion
12.
Pediatr Nephrol ; 31(12): 2201-2212, 2016 12.
Article in English | MEDLINE | ID: mdl-27011217

ABSTRACT

The oculocerebrorenal syndrome of Lowe is a rare X-linked multisystemic disorder characterized by the triad of congenital cataracts, intellectual disability, and proximal renal tubular dysfunction. Whereas the ocular manifestations and severe muscular hypotonia are the typical first diagnostic clues apparent at birth, the manifestations of incomplete renal Fanconi syndrome are often recognized only later in life. Other characteristic features are progressive severe growth retardation and behavioral problems, with tantrums. Many patients develop a debilitating arthropathy. Treatment is symptomatic, and the life span rarely exceeds 40 years. The causative oculocerebrorenal syndrome of Lowe gene (OCRL) encodes the inositol polyphosphate 5-phosphatase OCRL-1. OCRL variants have not only been found in classic Lowe syndrome, but also in patients with a predominantly renal phenotype classified as Dent disease type 2 (Dent-2). Recent data indicate that there is a phenotypic continuum between Dent-2 disease and Lowe syndrome, suggesting that there are individual differences in the ability to compensate for the loss of enzyme function. Extensive research has demonstrated that OCRL-1 is involved in multiple intracellular processes involving endocytic trafficking and actin skeleton dynamics. This explains the multi-organ manifestations of the disease. Still, the mechanisms underlying the wide phenotypic spectrum are poorly understood, and we are far from a causative therapy. In this review, we provide an update on clinical and molecular genetic findings in Lowe syndrome and the cellular and physiological functions of OCRL-1.


Subject(s)
Oculocerebrorenal Syndrome/therapy , Adolescent , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 11 , Humans , Infant , Infant, Newborn , Molecular Biology , Mutation , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , WAGR Syndrome
13.
Mol Biol Cell ; 27(1): 90-107, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26510499

ABSTRACT

Mutation of the inositol 5-phosphatase OCRL1 causes Lowe syndrome and Dent-2 disease. Loss of OCRL1 function perturbs several cellular processes, including membrane traffic, but the underlying mechanisms remain poorly defined. Here we show that OCRL1 is part of the membrane-trafficking machinery operating at the trans-Golgi network (TGN)/endosome interface. OCRL1 interacts via IPIP27A with the F-BAR protein pacsin 2. OCRL1 and IPIP27A localize to mannose 6-phosphate receptor (MPR)-containing trafficking intermediates, and loss of either protein leads to defective MPR carrier biogenesis at the TGN and endosomes. OCRL1 5-phosphatase activity, which is membrane curvature sensitive, is stimulated by IPIP27A-mediated engagement of OCRL1 with pacsin 2 and promotes scission of MPR-containing carriers. Our data indicate a role for OCRL1, via IPIP27A, in regulating the formation of pacsin 2-dependent trafficking intermediates and reveal a mechanism for coupling PtdIns(4,5)P2 hydrolysis with carrier biogenesis on endomembranes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Animals , COS Cells , Endocytosis , Endosomes/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , HEK293 Cells , HeLa Cells , Humans , Inositol Polyphosphate 5-Phosphatases , Nephrolithiasis/genetics , Nephrolithiasis/metabolism , Nephrolithiasis/pathology , Nerve Tissue Proteins/metabolism , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Phosphatidylinositols/biosynthesis , Phosphatidylinositols/metabolism , Protein Transport , Receptor, IGF Type 2/metabolism , trans-Golgi Network/metabolism
14.
Int Rev Cell Mol Biol ; 317: 331-47, 2015.
Article in English | MEDLINE | ID: mdl-26008789

ABSTRACT

Lowe syndrome is a lethal X-linked genetic disorder characterized by congenital cataracts, mental retardation, and kidney dysfunction. It is caused by mutations in the OCRL1 (oculocerebrorenal syndrome of Lowe) gene that encodes a phosphatidylinositol 5-phosphatase (EC 3.1.3.36). The gene product Ocrl1 has been linked to a multitude of functions due to the central role played by phosphoinositides in signaling. Moreover, this protein also has the ability to bind Rho GTPases, the master regulators of the actin cytoskeleton, and to interact with elements of the vesicle trafficking machinery. It is currently under investigation how deficiencies in Ocrl1 affect these different processes and contribute to patient symptoms. This chapter outlines the known physiological roles of Ocrl1 which might be relevant to the mechanism underlying Lowe syndrome.


Subject(s)
Cilia/physiology , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/metabolism , Animals , Humans
15.
Biochim Biophys Acta ; 1851(8): 1066-82, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25680866

ABSTRACT

Phosphoinositide (PIP) lipids regulate many aspects of cell function in the nervous system including receptor signalling, secretion, endocytosis, migration and survival. Levels of PIPs such as PI4P, PI(4,5)P2 and PI(3,4,5)P3 are normally tightly regulated by phosphoinositide kinases and phosphatases. Deregulation of these biochemical pathways leads to lipid imbalances, usually on intracellular endosomal membranes, and these changes have been linked to a number of major neurological diseases including Alzheimer's, Parkinson's, epilepsy, stroke, cancer and a range of rarer inherited disorders including brain overgrowth syndromes, Charcot-Marie-Tooth neuropathies and neurodevelopmental conditions such as Lowe's syndrome. This article analyses recent progress in this area and explains how PIP lipids are involved, to varying degrees, in almost every class of neurological disease. This article is part of a Special Issue entitled Brain Lipids.


Subject(s)
Alzheimer Disease/metabolism , Charcot-Marie-Tooth Disease/metabolism , Epilepsy/metabolism , Oculocerebrorenal Syndrome/metabolism , Parkinson Disease/metabolism , Phosphatidylinositols/metabolism , Stroke/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Epilepsy/genetics , Epilepsy/pathology , Gene Expression , Humans , Mutation , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Stroke/genetics , Stroke/pathology
16.
Biochim Biophys Acta ; 1851(6): 698-710, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25264170

ABSTRACT

Phosphoinositides (PIs) are a group of key signaling and structural lipid molecules involved in a myriad of cellular processes. PI phosphatases, together with PI kinases, are responsible for the conversion of PIs between distinctive phosphorylation states. PI phosphatases are a large collection of enzymes that are evolved from at least two disparate ancestors. One group is distantly related to endonucleases, which apply divalent metal ions for phosphoryl transfer. The other group is related to protein tyrosine phosphatases, which contain a highly conserved active site motif Cys-X5-Arg (CX5R). In this review, we focus on structural insights to illustrate current understandings of the molecular mechanisms of each PI phosphatase family, with emphasis on their structural basis for substrate specificity determinants and catalytic mechanisms. This article is part of a Special Issue entitled Phosphoinositides.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , PTEN Phosphohydrolase/chemistry , Phosphoric Monoester Hydrolases/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Bacteria/chemistry , Bacteria/enzymology , Bacterial Proteins/metabolism , Biocatalysis , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Humans , Inositol Polyphosphate 5-Phosphatases , Isoenzymes/chemistry , Isoenzymes/metabolism , Membrane Proteins/metabolism , Models, Molecular , Nephrolithiasis/enzymology , Nephrolithiasis/genetics , Nephrolithiasis/pathology , Oculocerebrorenal Syndrome/enzymology , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Substrate Specificity
17.
Elife ; 3: e02975, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25107275

ABSTRACT

Mutations in the inositol 5-phosphatase OCRL cause Lowe syndrome and Dent's disease. Although OCRL, a direct clathrin interactor, is recruited to late-stage clathrin-coated pits, clinical manifestations have been primarily attributed to intracellular sorting defects. Here we show that OCRL loss in Lowe syndrome patient fibroblasts impacts clathrin-mediated endocytosis and results in an endocytic defect. These cells exhibit an accumulation of clathrin-coated vesicles and an increase in U-shaped clathrin-coated pits, which may result from sequestration of coat components on uncoated vesicles. Endocytic vesicles that fail to lose their coat nucleate the majority of the numerous actin comets present in patient cells. SNX9, an adaptor that couples late-stage endocytic coated pits to actin polymerization and which we found to bind OCRL directly, remains associated with such vesicles. These results indicate that OCRL acts as an uncoating factor and that defects in clathrin-mediated endocytosis likely contribute to pathology in patients with OCRL mutations.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Fibroblasts/metabolism , Phosphoric Monoester Hydrolases/metabolism , Cells, Cultured , Clathrin-Coated Vesicles/metabolism , Clathrin-Coated Vesicles/ultrastructure , Coated Pits, Cell-Membrane/ultrastructure , Endocytosis/genetics , HEK293 Cells , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Electron , Microscopy, Fluorescence/methods , Mutation , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism , Oculocerebrorenal Syndrome/pathology , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/genetics , Protein Binding , Proteome/genetics , Proteome/metabolism , Proteomics/methods , RNA Interference , Sorting Nexins/genetics , Sorting Nexins/metabolism
18.
Turk J Pediatr ; 55(3): 331-4, 2013.
Article in English | MEDLINE | ID: mdl-24217083

ABSTRACT

Oculocerebrorenal syndrome of Lowe (OCRL) is a rare, X-linked disorder characterized by congenital cataracts, neonatal or infantile hypotonia, seizures, cognitive impairment, and renal tubular dysfunction. In this article, we report two maternal cousins with OCRL with a hemizygous p.Ala788Asp mutation in exon 22 of the OCRL gene. They presented with diverse features of selective proximal renal tubular defect and high serum levels of total cholesterol, low density lipoprotein cholesterol (LDL-C) and high density lipoprotein cholesterol (HDL-C).


Subject(s)
Dyslipidemias/complications , Kidney Tubules, Proximal/pathology , Oculocerebrorenal Syndrome/complications , Siblings , Cholesterol, HDL/blood , Cholesterol, LDL/blood , DNA/genetics , DNA Mutational Analysis , Dyslipidemias/blood , Humans , Infant , Infant, Newborn , Male , Mutation , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/genetics
19.
PLoS One ; 8(6): e66727, 2013.
Article in English | MEDLINE | ID: mdl-23805271

ABSTRACT

Inositol phosphatases are important regulators of cell signaling, polarity, and vesicular trafficking. Mutations in OCRL, an inositol polyphosphate 5-phosphatase, result in Oculocerebrorenal syndrome of Lowe, an X-linked recessive disorder that presents with congenital cataracts, glaucoma, renal dysfunction and mental retardation. INPP5B is a paralog of OCRL and shares similar structural domains. The roles of OCRL and INPP5B in the development of cataracts and glaucoma are not understood. Using ocular tissues, this study finds low levels of INPP5B present in human trabecular meshwork but high levels in murine trabecular meshwork. In contrast, OCRL is localized in the trabecular meshwork and Schlemm's canal endothelial cells in both human and murine eyes. In cultured human retinal pigmented epithelial cells, INPP5B was observed in the primary cilia. A functional role for INPP5B is revealed by defects in cilia formation in cells with silenced expression of INPP5B. This is further supported by the defective cilia formation in zebrafish Kupffer's vesicles and in cilia-dependent melanosome transport assays in inpp5b morphants. Taken together, this study indicates that OCRL and INPP5B are differentially expressed in the human and murine eyes, and play compensatory roles in cilia development.


Subject(s)
Cilia/metabolism , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/metabolism , Animals , Cells, Cultured , Cilia/pathology , Eye/metabolism , Eye/pathology , Humans , Immunohistochemistry , Mice , Microscopy, Fluorescence , Morpholinos/metabolism , Oculocerebrorenal Syndrome/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...