Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Physiol ; 602(17): 4291-4307, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39106251

ABSTRACT

ClC-K/barttin channels are involved in the transepithelial transport of chloride in the kidney and inner ear. Their physiological role is crucial in humans because mutations in CLCNKB or BSND, encoding ClC-Kb and barttin, cause Bartter's syndrome types III and IV, respectively. In vitro experiments have shown that an amino acid change in a proline-tyrosine motif in the C-terminus of barttin stimulates ClC-K currents. The molecular mechanism of this enhancement and whether this potentiation has any in vivo relevance remains unknown. We performed electrophysiological and biochemical experiments in Xenopus oocytes and kidney cells co-expressing ClC-K and barttin constructs. We demonstrated that barttin possesses a YxxØ motif and, when mutated, increases ClC-K plasma membrane stability, resulting in larger currents. To address the impact of mutating this motif in kidney physiology, we generated a knock-in mouse. Comparing wild-type (WT) and knock-in mice under a standard diet, we could not observe any difference in ClC-K and barttin protein levels or localization, either in urinary or plasma parameters. However, under a high-sodium low-potassium diet, known to induce hyperplasia of distal convoluted tubules, knock-in mice exhibit reduced hyperplasia compared to WT mice. In summary, our in vitro and in vivo studies demonstrate that the previously identified PY motif is indeed an endocytic YxxØ motif in which mutations cause a gain of function of the channel. KEY POINTS: It is revealed by mutagenesis and functional experiments that a previously identified proline-tyrosine motif regulating ClC-K plasma membrane levels is indeed an endocytic YxxØ motif. Biochemical characterization of mutants in the YxxØ motif in Xenopus oocytes and human embryonic kidney cells indicates that mutants showed increased plasma membrane levels as a result of an increased stability, resulting in higher function of ClC-K channels. Mutation of this motif does not affect barttin protein expression and subcellular localization in vivo. Knock-in mice with a mutation in this motif, under conditions of a high-sodium low-potassium diet, exhibit less hyperplasia in the distal convoluted tubule than wild-type animals, indicating a gain of function of the channel in vivo.


Subject(s)
Chloride Channels , Endocytosis , Xenopus laevis , Animals , Chloride Channels/genetics , Chloride Channels/metabolism , Endocytosis/physiology , Mice , Kidney Tubules, Distal/metabolism , Hyperplasia , Humans , Female , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Mice, Inbred C57BL , HEK293 Cells , Oocytes/metabolism , Anion Transport Proteins
2.
Hum Mol Genet ; 30(17): 1649-1665, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34100078

ABSTRACT

Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events.


Subject(s)
Cysts/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuron-Glia/genetics , Cell Adhesion Molecules, Neuron-Glia/metabolism , Cell Cycle Proteins/genetics , Chloride Channels/genetics , Cysts/metabolism , HEK293 Cells , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System Malformations/metabolism , Protein Transport , Receptors, G-Protein-Coupled/metabolism
3.
Int J Mol Sci ; 24(24)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38138969

ABSTRACT

More than 20 years have passed since the identification of SLC3A1 and SLC7A9 as causative genes for cystinuria. However, cystinuria patients exhibit significant variability in the age of lithiasis onset, recurrence, and response to treatment, suggesting the presence of modulatory factors influencing cystinuria severity. In 2016, a second renal cystine transporter, AGT1, encoded by the SLC7A13 gene, was discovered. Although it was discarded as a causative gene for cystinuria, its possible effect as a modulatory gene remains unexplored. Thus, we analyzed its function in mouse models of cystinuria, screened the SLC7A13 gene in 34 patients with different lithiasic phenotypes, and functionally characterized the identified variants. Mice results showed that AGT1/rBAT may have a protective role against cystine lithiasis. In addition, among the four missense variants detected in patients, two exhibited a 25% impairment in AGT1/rBAT transport. However, no correlation between SLC7A13 genotypes and lithiasis phenotypes was observed in patients, probably because these variants were found in heterozygous states. In conclusion, our results, consistent with a previous study, suggest that AGT1/rBAT does not have a relevant effect on cystinuria patients, although an impact in patients carrying homozygous pathogenic variants cannot be discarded.


Subject(s)
Cystinuria , Lithiasis , Humans , Animals , Mice , Cystinuria/genetics , Cystinuria/pathology , Lithiasis/complications , Cystine , Retrospective Studies , Kidney/pathology
4.
Sensors (Basel) ; 20(23)2020 Nov 24.
Article in English | MEDLINE | ID: mdl-33255308

ABSTRACT

Several pathologies have a direct impact on society, causing public health problems. Pulmonary diseases such as Chronic obstructive pulmonary disease (COPD) are already the third leading cause of death in the world, leaving tuberculosis at ninth with 1.7 million deaths and over 10.4 million new occurrences. The detection of lung regions in images is a classic medical challenge. Studies show that computational methods contribute significantly to the medical diagnosis of lung pathologies by Computerized Tomography (CT), as well as through Internet of Things (IoT) methods based in the context on the health of things. The present work proposes a new model based on IoT for classification and segmentation of pulmonary CT images, applying the transfer learning technique in deep learning methods combined with Parzen's probability density. The proposed model uses an Application Programming Interface (API) based on the Internet of Medical Things to classify lung images. The approach was very effective, with results above 98% accuracy for classification in pulmonary images. Then the model proceeds to the lung segmentation stage using the Mask R-CNN network to create a pulmonary map and use fine-tuning to find the pulmonary borders on the CT image. The experiment was a success, the proposed method performed better than other works in the literature, reaching high segmentation metrics values such as accuracy of 98.34%. Besides reaching 5.43 s in segmentation time and overcoming other transfer learning models, our methodology stands out among the others because it is fully automatic. The proposed approach has simplified the segmentation process using transfer learning. It has introduced a faster and more effective method for better-performing lung segmentation, making our model fully automatic and robust.


Subject(s)
Deep Learning , Internet of Things , Tomography, X-Ray Computed , Image Processing, Computer-Assisted , Lung/diagnostic imaging
5.
Hum Mol Genet ; 26(13): 2436-2450, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28398517

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy caused by mutations in either MLC1 or GLIALCAM. GlialCAM is necessary for the correct targeting of MLC1, but also for the targeting of the Cl- channel ClC-2. Furthermore, GlialCAM modifies ClC-2 functional properties in vitro. However, in vivo studies in GlialCAM-/- mice have shown that the modification of ClC-2 activity only occurs in oligodendrocytes, despite GlialCAM and ClC-2 being expressed in astrocytes. Thus, the relationship between GlialCAM, MLC1 and ClC-2 in astrocytes is unknown. Here, we show that GlialCAM, ClC-2 and MLC1 can form a ternary complex in cultured astrocytes, but only under depolarizing conditions. We also provide biochemical evidences that this ternary complex exists in vivo. The formation of this complex changes ClC-2 localization in the membrane and its functional properties. ClC-2 association with GlialCAM/MLC1 depends on calcium flux through L-type calcium channels and activation of calcium-dependent calpain proteases. Based on these studies, we propose that the chloride influx mediated by GlialCAM/MLC1/ClC-2 in astrocytes may be needed to compensate an excess of potassium, as occurs in conditions of high neuronal activity. We suggest that a defect in this compensation may contribute to the pathogenesis of MLC disease.


Subject(s)
Cell Adhesion Molecules, Neuron-Glia/metabolism , Cysts/metabolism , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Brain Diseases/pathology , CLC-2 Chloride Channels , Calcium Channels, L-Type/genetics , Chloride Channels , Cysts/genetics , HEK293 Cells , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/genetics , Humans , Membrane Proteins/genetics , Mice , Protein Transport/genetics
6.
Proc Natl Acad Sci U S A ; 113(3): 775-80, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26739563

ABSTRACT

Heterodimeric amino acid transporters play crucial roles in epithelial transport, as well as in cellular nutrition. Among them, the heterodimer of a membrane protein b(0,+)AT/SLC7A9 and its auxiliary subunit rBAT/SLC3A1 is responsible for cystine reabsorption in renal proximal tubules. The mutations in either subunit cause cystinuria, an inherited amino aciduria with impaired renal reabsorption of cystine and dibasic amino acids. However, an unsolved paradox is that rBAT is highly expressed in the S3 segment, the late proximal tubules, whereas b(0,+)AT expression is highest in the S1 segment, the early proximal tubules, so that the presence of an unknown partner of rBAT in the S3 segment has been proposed. In this study, by means of coimmunoprecipitation followed by mass spectrometry, we have found that a membrane protein AGT1/SLC7A13 is the second partner of rBAT. AGT1 is localized in the apical membrane of the S3 segment, where it forms a heterodimer with rBAT. Depletion of rBAT in mice eliminates the expression of AGT1 in the renal apical membrane. We have reconstituted the purified AGT1-rBAT heterodimer into proteoliposomes and showed that AGT1 transports cystine, aspartate, and glutamate. In the apical membrane of the S3 segment, AGT1 is suggested to locate itself in close proximity to sodium-dependent acidic amino acid transporter EAAC1 for efficient functional coupling. EAAC1 is proposed to take up aspartate and glutamate released into luminal fluid by AGT1 due to its countertransport so that preventing the urinary loss of aspartate and glutamate. Taken all together, AGT1 is the long-postulated second cystine transporter in the S3 segment of proximal tubules and a possible candidate to be involved in isolated cystinuria.


Subject(s)
Amino Acid Transport Systems, Basic/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Amino Acid Transport Systems/metabolism , Cell Membrane/metabolism , Cystinuria/metabolism , Kidney Tubules, Proximal/metabolism , Amino Acid Sequence , Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/genetics , Animals , Antibodies/metabolism , Blotting, Western , Excitatory Amino Acid Transporter 3/metabolism , Female , HEK293 Cells , Humans , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Male , Mice, Inbred C57BL , Molecular Sequence Data , Protein Binding , Protein Multimerization , Proteolipids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics
7.
J Am Soc Nephrol ; 29(6): 1624-1635, 2018 06.
Article in English | MEDLINE | ID: mdl-29610403

ABSTRACT

Background Reabsorption of amino acids (AAs) across the renal proximal tubule is crucial for intracellular and whole organism AA homeostasis. Although the luminal transport step is well understood, with several diseases caused by dysregulation of this process, the basolateral transport step is not understood. In humans, only cationic aminoaciduria due to malfunction of the basolateral transporter y+LAT1/CD98hc (SLC7A7/SLC3A2), which mediates the export of cationic AAs, has been described. Thus, the physiologic roles of basolateral transporters of neutral AAs, such as the antiporter LAT2/CD98hc (SLC7A8/SLC3A2), a heterodimer that exports most neutral AAs, and the uniporter TAT1 (SLC16A10), which exports only aromatic AAs, remain unclear. Functional cooperation between TAT1 and LAT2/CD98hc has been suggested by in vitro studies but has not been evaluated in vivoMethods To study the functional relationship of TAT1 and LAT2/CD98hc in vivo, we generated a double-knockout mouse model lacking TAT1 and LAT2, the catalytic subunit of LAT2/CD98hc (dKO LAT2-TAT1 mice).Results Compared with mice lacking only TAT1 or LAT2, dKO LAT2-TAT1 mice lost larger amounts of aromatic and other neutral AAs in their urine due to a tubular reabsorption defect. Notably, dKO mice also displayed decreased tubular reabsorption of cationic AAs and increased expression of y+LAT1/CD98hc.Conclusions The LAT2/CD98hc and TAT1 transporters functionally cooperate in vivo, and y+LAT1/CD98hc may compensate for the loss of LAT2/CD98hc and TAT1, functioning as a neutral AA exporter at the expense of some urinary loss of cationic AAs. Cooperative and compensatory mechanisms of AA transporters may explain the lack of basolateral neutral aminoacidurias in humans.


Subject(s)
Amino Acid Transport System y+/genetics , Amino Acid Transport Systems, Neutral/genetics , Amino Acids, Neutral/metabolism , Fusion Regulatory Protein 1, Light Chains/genetics , Renal Reabsorption , Amino Acid Transport System y+/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids, Neutral/urine , Animals , Female , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Light Chains/metabolism , Kidney Tubules/physiology , Male , Mice, Knockout
8.
Neurobiol Dis ; 119: 88-99, 2018 11.
Article in English | MEDLINE | ID: mdl-30076890

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy caused by mutations in either MLC1 or GLIALCAM genes. Previous work indicated that chloride currents mediated by the volume-regulated anion channel (VRAC) and ClC-2 channels were affected in astrocytes deficient in either Mlc1 or Glialcam. ClC-2 forms a ternary complex with GlialCAM and MLC1. LRRC8 proteins have been identified recently as the molecular components of VRAC, but the relationship between MLC and LRRC8 proteins is unknown. Here, we first demonstrate that LRRC8 and MLC1 are functionally linked, as MLC1 cannot potentiate VRAC currents when LRRC8A, the main subunit of VRAC, is knocked down. We determine that LRRC8A and MLC1 do not co-localize or interact and, in Xenopus oocytes, MLC1 does not potentiate LRRC8-mediated VRAC currents, indicating that VRAC modulation in astrocytes by MLC1 may be indirect. Investigating the mechanism of modulation, we find that a lack of MLC1 does not influence either mRNA or total and plasma membrane protein levels of LRRC8A; and neither does it affect LRRC8A subcellular localization. In agreement with recent results that indicated that overexpression of MLC1 decreases the phosphorylation of extracellular signal-regulated kinases (ERK), we find that astrocytes lacking MLC1 show an increase in ERK phosphorylation. In astrocytes with reduced or increased levels of MLC1 we observe changes in the phosphorylation state of the VRAC subunit LRRC8C. Our results thus reinforce previous suggestions that indicated that GlialCAM/MLC1 might modify signal transduction pathways that influence the activity of different proteins, such as VRAC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , Cysts/metabolism , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Astrocytes/chemistry , Astrocytes/pathology , Cell Cycle Proteins , Cells, Cultured , Cysts/pathology , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Membrane Proteins/analysis , Membrane Proteins/genetics , Proteins/analysis , Proteins/genetics , Rats , Xenopus
9.
Hum Mutat ; 38(7): 764-777, 2017 07.
Article in English | MEDLINE | ID: mdl-28432734

ABSTRACT

We developed a variant database for diabetes syndrome genes, using the Leiden Open Variation Database platform, containing observed phenotypes matched to the genetic variations. We populated it with 628 published disease-associated variants (December 2016) for: WFS1 (n = 309), CISD2 (n = 3), ALMS1 (n = 268), and SLC19A2 (n = 48) for Wolfram type 1, Wolfram type 2, Alström, and Thiamine-responsive megaloblastic anemia syndromes, respectively; and included 23 previously unpublished novel germline variants in WFS1 and 17 variants in ALMS1. We then investigated genotype-phenotype relations for the WFS1 gene. The presence of biallelic loss-of-function variants predicted Wolfram syndrome defined by insulin-dependent diabetes and optic atrophy, with a sensitivity of 79% (95% CI 75%-83%) and specificity of 92% (83%-97%). The presence of minor loss-of-function variants in WFS1 predicted isolated diabetes, isolated deafness, or isolated congenital cataracts without development of the full syndrome (sensitivity 100% [93%-100%]; specificity 78% [73%-82%]). The ability to provide a prognostic prediction based on genotype will lead to improvements in patient care and counseling. The development of the database as a repository for monogenic diabetes gene variants will allow prognostic predictions for other diabetes syndromes as next-generation sequencing expands the repertoire of genotypes and phenotypes. The database is publicly available online at https://lovd.euro-wabb.org.


Subject(s)
Anemia, Megaloblastic/genetics , Databases, Genetic , Diabetes Mellitus/genetics , Hearing Loss, Sensorineural/genetics , Thiamine Deficiency/congenital , Wolfram Syndrome/genetics , Adolescent , Adult , Child , Child, Preschool , Exons , Family Health , Female , Genetic Association Studies , Genetic Variation , Genotype , Homozygote , Humans , Male , Phenotype , Prognosis , Sensitivity and Specificity , Thiamine Deficiency/genetics , Young Adult
10.
Hum Mol Genet ; 23(19): 5069-86, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24824219

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a leukodystrophy characterized by myelin vacuolization and caused by mutations in MLC1 or GLIALCAM. Patients with recessive mutations in either MLC1 or GLIALCAM show the same clinical phenotype. It has been shown that GLIALCAM is necessary for the correct targeting of MLC1 to the membrane at cell junctions, but its own localization was independent of MLC1 in vitro. However, recent studies in Mlc1(-/-) mice have shown that GlialCAM is mislocalized in glial cells. In order to investigate whether the relationship between Mlc1 and GlialCAM is species-specific, we first identified MLC-related genes in zebrafish and generated an mlc1(-/-) zebrafish. We have characterized mlc1(-/-) zebrafish both functionally and histologically and compared the phenotype with that of the Mlc1(-/-) mice. In mlc1(-/-) zebrafish, as in Mlc1(-/-) mice, Glialcam is mislocalized. Re-examination of a brain biopsy from an MLC patient indicates that GLIALCAM is also mislocalized in Bergmann glia in the cerebellum. In vitro, impaired localization of GlialCAM was observed in astrocyte cultures from Mlc1(-/-) mouse only in the presence of elevated potassium levels, which mimics neuronal activity. In summary, here we demonstrate an evolutionary conserved role for MLC1 in regulating glial surface levels of GLIALCAM, and this interrelationship explains why patients with mutations in either gene (MLC1 or GLIALCAM) share the same clinical phenotype.


Subject(s)
Cysts/metabolism , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Membrane Proteins/metabolism , Neuroglia/metabolism , Proteins/metabolism , Animals , Animals, Genetically Modified , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Cycle Proteins , Cell Line , Cell Membrane/metabolism , Cysts/genetics , Disease Models, Animal , Ependyma/cytology , Ependyma/metabolism , Ependyma/ultrastructure , Gene Expression , Genotype , Hereditary Central Nervous System Demyelinating Diseases/genetics , Humans , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Phenotype , Protein Transport , Proteins/genetics , Retina/metabolism , Voltage-Dependent Anion Channels/metabolism , Zebrafish/genetics , Zebrafish/metabolism
11.
Hum Mol Genet ; 22(21): 4405-16, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23793458

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy caused by mutations in either MLC1 or GLIALCAM genes and is associated with myelin and astrocyte vacuolation. It has been suggested that MLC is caused by impaired cell volume regulation as a result of defective activation of astrocytic volume-regulated anion currents (VRAC). GlialCAM brings MLC1 and the ClC-2 Cl(-) channel to cell-cell junctions, even though the role of ClC-2 in MLC disease remains incompletely understood. To gain insights into the biological role of GlialCAM in the pathogenesis of MLC disease, here we analyzed the gain- and loss-of-function phenotypes of GlialCAM in Hela cells and primary astrocytes, focusing on its interaction with the MLC1 protein. Unexpectedly, GlialCAM ablation provoked intracellular accumulation and reduced expression of MLC1 at the plasma membrane. Conversely, over-expression of GlialCAM increased the cellular stability of mutant MLC1 variants. Reduction in GlialCAM expression resulted in defective activation of VRAC and augmented vacuolation, phenocopying MLC1 mutations. Importantly, over-expression of GlialCAM together with MLC1 containing MLC-related mutations was able to reactivate VRAC currents and to reverse the vacuolation caused in the presence of mutant MLC1. These results indicate a previously unrecognized role of GlialCAM in facilitating the biosynthetic maturation and cell surface expression of MLC1, and suggest that pharmacological strategies aimed to increase surface expression of MLC1 and/or VRAC activity may be beneficial for MLC patients.


Subject(s)
Astrocytes/physiology , Chloride Channels/physiology , Cysts/physiopathology , Hereditary Central Nervous System Demyelinating Diseases/physiopathology , Proteins/genetics , Proteins/metabolism , Animals , Astrocytes/ultrastructure , CLC-2 Chloride Channels , Cell Cycle Proteins , Cysts/genetics , Genetic Variation , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Phenotype , Protein Stability , RNA Interference , Rats , Vacuoles/physiology
12.
Hum Mutat ; 35(4): 470-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449431

ABSTRACT

Inactivating mutations in the BCKDK gene, which codes for the kinase responsible for the negative regulation of the branched-chain α-keto acid dehydrogenase complex (BCKD), have recently been associated with a form of autism in three families. In this work, two novel exonic BCKDK mutations, c.520C>G/p.R174G and c.1166T>C/p.L389P, were identified at the homozygous state in two unrelated children with persistently reduced body fluid levels of branched-chain amino acids (BCAAs), developmental delay, microcephaly, and neurobehavioral abnormalities. Functional analysis of the mutations confirmed the missense character of the c.1166T>C change and showed a splicing defect r.[520c>g;521_543del]/p.R174Gfs1*, for c.520C>G due to the presence of a new donor splice site. Mutation p.L389P showed total loss of kinase activity. Moreover, patient-derived fibroblasts showed undetectable (p.R174Gfs1*) or barely detectable (p.L389P) levels of BCKDK protein and its phosphorylated substrate (phospho-E1α), resulting in increased BCKD activity and the very rapid BCAA catabolism manifested by the patients' clinical phenotype. Based on these results, a protein-rich diet plus oral BCAA supplementation was implemented in the patient homozygous for p.R174Gfs1*. This treatment normalized plasma BCAA levels and improved growth, developmental and behavioral variables. Our results demonstrate that BCKDK mutations can result in neurobehavioral deficits in humans and support the rationale for dietary intervention.


Subject(s)
Developmental Disabilities/genetics , Nervous System Diseases/genetics , Protein Kinases/genetics , Amino Acids, Branched-Chain/administration & dosage , Amino Acids, Branched-Chain/blood , Developmental Disabilities/diet therapy , Fibroblasts/enzymology , Humans , Male , Mutation, Missense , Nervous System Diseases/diet therapy , Pediatrics , Protein Kinases/deficiency
13.
Biochim Biophys Acta ; 1833(10): 2311-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23732701

ABSTRACT

Cyclosporine A and FK506 produce immunosuppression by blocking calcineurin phosphatase activity and consequently activation of cytosolic Nuclear Factor of Activated T-cell (NFATc) transcription factor. Due to the chronic toxicity associated with their administration, the development of more specific immunosuppressants is currently an important unmet medical need. In this context, an immunosuppressant peptide derived from the CIC motif of the human Regulators of Calcineurin (RCAN) proteins has been shown to inhibit NFATc signaling without affecting general phosphatase activity of calcineurin. Here we show that protein kinase CK2 phosphorylates a conserved serine residue within the CIC motif of vertebrate RCANs, which increases its affinity for calcineurin and consequently its inhibition of NFATc-dependent gene expression in activated T-cells. Molecular modeling studies have led us to identify a positively charged interaction site on the surface of calcineurin where the phosphorylated serine residue of the CIC motif would normally locate. Finally, we have also identified RCAN3 as a new phosphoprotein with multiple phosphorylation sites. Therefore, our findings reveal for the first time a novel molecular mechanism underlying the regulation of calcineurin-NFATc signaling by means of phosphorylation of the CIC motif of RCAN proteins. The knowledge of how RCAN proteins modulate the calcineurin-NFATc pathway paves the way for the development of potent novel selective immunosuppressant drugs.


Subject(s)
Calcineurin/metabolism , Casein Kinase II/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Amino Acid Sequence , Blotting, Western , Calcineurin/genetics , Casein Kinase II/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Circular Dichroism , DNA-Binding Proteins , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Luciferases/metabolism , Molecular Sequence Data , Muscle Proteins/genetics , NFATC Transcription Factors/genetics , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction
14.
Am J Hum Genet ; 88(4): 422-32, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21419380

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a leukodystrophy characterized by early-onset macrocephaly and delayed-onset neurological deterioration. Recessive MLC1 mutations are observed in 75% of patients with MLC. Genetic-linkage studies failed to identify another gene. We recently showed that some patients without MLC1 mutations display the classical phenotype; others improve or become normal but retain macrocephaly. To find another MLC-related gene, we used quantitative proteomic analysis of affinity-purified MLC1 as an alternative approach and found that GlialCAM, an IgG-like cell adhesion molecule that is also called HepaCAM and is encoded by HEPACAM, is a direct MLC1-binding partner. Analysis of 40 MLC patients without MLC1 mutations revealed multiple different HEPACAM mutations. Ten patients with the classical, deteriorating phenotype had two mutations, and 18 patients with the improving phenotype had one mutation. Most parents with a single mutation had macrocephaly, indicating dominant inheritance. In some families with dominant HEPACAM mutations, the clinical picture and magnetic resonance imaging normalized, indicating that HEPACAM mutations can cause benign familial macrocephaly. In other families with dominant HEPACAM mutations, patients had macrocephaly and mental retardation with or without autism. Further experiments demonstrated that GlialCAM and MLC1 both localize in axons and colocalize in junctions between astrocytes. GlialCAM is additionally located in myelin. Mutant GlialCAM disrupts the localization of MLC1-GlialCAM complexes in astrocytic junctions in a manner reflecting the mode of inheritance. In conclusion, GlialCAM is required for proper localization of MLC1. HEPACAM is the second gene found to be mutated in MLC. Dominant HEPACAM mutations can cause either macrocephaly and mental retardation with or without autism or benign familial macrocephaly.


Subject(s)
Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Intellectual Disability/genetics , Megalencephaly/genetics , Mutation , Proteins/genetics , Amino Acid Sequence , Animals , Autistic Disorder/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Cycle Proteins , Cells, Cultured , Cysts/genetics , Cysts/metabolism , Genes, Dominant , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/metabolism , Humans , Intellectual Disability/metabolism , Megalencephaly/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs/genetics , Proteins/metabolism , Rats , Sequence Homology, Amino Acid
15.
Am J Physiol Renal Physiol ; 305(12): F1645-55, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24107421

ABSTRACT

Cystinuria is an autosomal recessive disease caused by mutations in SLC3A1 (rBAT) and SLC7A9 (b(0,+)AT). Gene targeting of the catalytic subunit (Slc7a9) in mice leads to excessive excretion of cystine, lysine, arginine, and ornithine. Here, we studied this non-type I cystinuria mouse model using gene expression analysis, Western blotting, clearance, and brush-border membrane vesicle (BBMV) uptake experiments to further characterize the renal and intestinal consequences of losing Slc7a9 function. The electrogenic and BBMV flux studies in the intestine suggested that arginine and ornithine are transported via other routes apart from system b(0,+). No remarkable gene expression changes were observed in other amino acid transporters and the peptide transporters in the intestine and kidney. Furthermore, the glomerular filtration rate (GFR) was reduced by 30% in knockout animals compared with wild-type animals. The fractional excretion of arginine was increased as expected (∼100%), but fractional excretions of lysine (∼35%), ornithine (∼16%), and cystine (∼11%) were less affected. Loss of function of b(0,+)AT reduced transport of cystine and arginine in renal BBMVs and completely abolished the exchanger activity of dibasic amino acids with neutral amino acids. In conclusion, loss of Slc7a9 function decreases the GFR and increases the excretion of several amino acids to a lesser extent than expected with no clear regulation at the mRNA and protein level of alternative transporters and no increased renal epithelial uptake. These observations indicate that transporters located in distal segments of the kidney and/or metabolic pathways may partially compensate for Slc7a9 loss of function.


Subject(s)
Amino Acid Transport Systems, Basic/deficiency , Amino Acids, Diamino/metabolism , Cystine/metabolism , Cystinuria/metabolism , Amino Acid Transport Systems, Basic/genetics , Animals , Cystinuria/physiopathology , Disease Models, Animal , Glomerular Filtration Rate/physiology , Kidney/metabolism , Kidney/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Hum Mol Genet ; 20(16): 3266-77, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21624973

ABSTRACT

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare leukodystrophy caused by mutations in MLC1 or GLIALCAM. The GLIALCAM gene product functions as an MLC1 beta-subunit. We aim to further clarify the molecular mechanisms of MLC caused by mutations in MLC1 or GLIALCAM. For this purpose, we analyzed a human post-mortem brain obtained from an MLC patient, who was homozygous for a missense mutation (S69L) in MLC1. We showed that this mutation affects the stability of MLC1 in vitro and reduces MLC1 protein levels in the brain to almost undetectable. However, the amount of GlialCAM and its localization were nearly unaffected, indicating that MLC1 is not necessary for GlialCAM expression or targeting. These findings were supported by experiments in primary astrocytes and in heterologous cells. In addition, we demonstrated that MLC1 and GlialCAM form homo- and hetero-complexes and that MLC-causing mutations in GLIALCAM mainly reduce the formation of GlialCAM homo-complexes, leading to a defect in the trafficking of GlialCAM alone to cell junctions. GLIALCAM mutations also affect the trafficking of its associated molecule MLC1, explaining why GLIALCAM and MLC1 mutations lead to the same disease: MLC.


Subject(s)
Cysts/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Membrane Proteins/genetics , Mutation/genetics , Proteins/genetics , Adult , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cell Cycle Proteins , Cysts/pathology , Fatal Outcome , Female , HEK293 Cells , HeLa Cells , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Middle Aged , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Transport , RNA Interference , Rats , Transfection
17.
Genet Med ; 15(7): 497-506, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23429432

ABSTRACT

PURPOSE: Wolfram syndrome is a degenerative, recessive rare disease with an onset in childhood. It is caused by mutations in WFS1 or CISD2 genes. More than 200 different variations in WFS1 have been described in patients with Wolfram syndrome, which complicates the establishment of clear genotype-phenotype correlation. The purpose of this study was to elucidate the role of WFS1 mutations and update the natural history of the disease. METHODS: This study analyzed clinical and genetic data of 412 patients with Wolfram syndrome published in the last 15 years. RESULTS: (i) 15% of published patients do not fulfill the current -inclusion criterion; (ii) genotypic prevalence differences may exist among countries; (iii) diabetes mellitus and optic atrophy might not be the first two clinical features in some patients; (iv) mutations are nonuniformly distributed in WFS1; (v) age at onset of diabetes mellitus, hearing defects, and diabetes insipidus may depend on the patient's genotypic class; and (vi) disease progression rate might depend on genotypic class. CONCLUSION: New genotype-phenotype correlations were established, disease progression rate for the general population and for the genotypic classes has been calculated, and new diagnostic criteria have been proposed. The conclusions raised could be important for patient management and counseling as well as for the development of treatments for Wolfram syndrome.


Subject(s)
Membrane Proteins/genetics , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Age of Onset , Diabetes Insipidus/genetics , Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Genetic Association Studies , Humans , Membrane Proteins/metabolism , Mutation , Optic Atrophy/genetics , Wolfram Syndrome/classification , Wolfram Syndrome/etiology
18.
BMC Pediatr ; 13: 130, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23981649

ABSTRACT

BACKGROUND: Wolfram, Alström and Bardet-Biedl (WABB) syndromes are rare diseases with overlapping features of multiple sensory and metabolic impairments, including diabetes mellitus, which have caused diagnostic confusion. There are as yet no specific treatments available, little or no access to well characterized cohorts of patients, and limited information on the natural history of the diseases. We aim to establish a Europe-wide registry for these diseases to inform patient care and research. METHODS: EURO-WABB is an international multicenter large-scale observational study capturing longitudinal clinical and outcome data for patients with WABB diagnoses. Three hundred participants will be recruited over 3 years from different sites throughout Europe. Comprehensive clinical, genetic and patient experience data will be collated into an anonymized disease registry. Data collection will be web-based, and forms part of the project's Virtual Research and Information Environment (VRIE). Participants who haven't undergone genetic diagnostic testing for their condition will be able to do so via the project. CONCLUSIONS: The registry data will be used to increase the understanding of the natural history of WABB diseases, to serve as an evidence base for clinical management, and to aid the identification of opportunities for intervention to stop or delay the progress of the disease. The detailed clinical characterisation will allow inclusion of patients into studies of novel treatment interventions, including targeted interventions in small scale open label studies; and enrolment into multi-national clinical trials. The registry will also support wider access to genetic testing, and encourage international collaborations for patient benefit.


Subject(s)
Alstrom Syndrome , Bardet-Biedl Syndrome , Rare Diseases , Registries , Wolfram Syndrome , Adolescent , Adult , Child , Child, Preschool , Databases as Topic , European Union , Female , Genetic Testing , Humans , Infant , International Cooperation , Male , Research Design
19.
Front Genet ; 14: 1198171, 2023.
Article in English | MEDLINE | ID: mdl-37415600

ABSTRACT

Objective: Wolfram syndrome (WFS) is an autosomal recessive disorder associated with juvenile-onset diabetes mellitus, optic atrophy, diabetes insipidus, and sensorineural hearing loss. We sought to elucidate the relationship between genotypic and phenotypic presentations of Wolfram syndrome which would assist clinicians in classifying the severity and prognosis of Wolfram syndrome more accurately. Approach: Patient data from the Washington University International Registry and Clinical Study for Wolfram Syndrome and patient case reports were analyzed to select for patients with two recessive mutations in the WFS1 gene. Mutations were classified as being either nonsense/frameshift variants or missense/in-frame insertion/deletion variants. Missense/in-frame variants were further classified as transmembrane or non-transmembrane based on whether they affected amino acid residues predicted to be in transmembrane domains of WFS1. Statistical analysis was performed using Wilcoxon rank-sum tests with multiple test adjustment applied via the Bonferonni correction. Results: A greater number of genotype variants correlated with earlier onset and a more severe presentation of Wolfram syndrome. Secondly, non-sense and frameshift variants had more severe phenotypic presentations than missense variants, as evidenced by diabetes mellitus and optic atrophy emerging significantly earlier in patients with two nonsense/frameshift variants compared with zero or one nonsense/frameshift variants. In addition, the number of transmembrane in-frame variants demonstrated a statistically significant dose-effect on age of onset of diabetes mellitus and optic atrophy among patients with either one or two in-frame variants. Summary/Conclusion: The results contribute to our current understanding of the genotype-phenotype relationship of Wolfram syndrome, suggesting that alterations in coding sequences result in significant changes in the presentation and severity of Wolfram. The impact of these findings is significant, as the results will aid clinicians in predicting more accurate prognoses and pave the way for personalized treatments for Wolfram syndrome.

20.
medRxiv ; 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36824811

ABSTRACT

Objective: Wolfram syndrome (WFS) is an autosomal recessive disorder associated with juvenile-onset diabetes mellitus, optic atrophy, diabetes insipidus, and sensorineural hearing loss. We sought to elucidate the relationship between genotypic and phenotypic presentations of Wolfram syndrome which would assist clinicians in classifying the severity and prognosis of Wolfram syndrome more accurately. Approach: Patient data from the Washington University International Registry and Clinical Study for Wolfram Syndrome and patient case reports were analyzed to select for patients with two recessive mutations in the WFS1 gene. Mutations were classified as being either nonsense/frameshift variants or missense/in-frame insertion/deletion variants and statistical analysis was performed using unpaired and paired t-tests and one- and two-way ANOVA with Tukey's or Dunnett's tests. Results: A greater number of genotype variants correlated with earlier onset and a more severe presentation of Wolfram syndrome. Secondly, non-sense and frameshift variants had more severe phenotypic presentations than missense variants, as evidenced by optic atrophy emerging significantly earlier in patients with 2 nonsense/frameshift alleles compared with 0 missense transmembrane variants. In addition, the number of transmembrane in-frame variants demonstrated a statistically significant dose-effect on age of onset of diabetes mellitus and optic atrophy. Summary / Conclusions: The results contribute to our current understanding of the genotype-phenotype relationship of Wolfram syndrome, suggesting that alterations in coding sequences result in significant changes in the presentation and severity of Wolfram. The impact of these findings is significant, as the results will aid clinicians in predicting more accurate prognoses and pave the way for personalized treatments for Wolfram syndrome.

SELECTION OF CITATIONS
SEARCH DETAIL