Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(3): 167, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35233680

RESUMEN

The cellular defense mechanisms against cumulative endo-lysosomal stress remain incompletely understood. Here, we identify Ubr1 as a protein quality control (QC) E3 ubiquitin-ligase that counteracts proteostasis stresses by facilitating endosomal cargo-selective autophagy for lysosomal degradation. Astrocyte regulatory cluster membrane protein MLC1 mutations cause endosomal compartment stress by fusion and enlargement. Partial lysosomal clearance of mutant endosomal MLC1 is accomplished by the endosomal QC ubiquitin ligases, CHIP and Ubr1 via ESCRT-dependent route. As a consequence of the endosomal stress, a supportive QC mechanism, dependent on both Ubr1 and SQSTM1/p62 activities, targets ubiquitinated and arginylated MLC1 mutants for selective endosomal autophagy (endophagy). This QC pathway is also activated for arginylated Ubr1-SQSTM1/p62 autophagy cargoes during cytosolic Ca2+-assault. Conversely, the loss of Ubr1 and/or arginylation elicited endosomal compartment stress. These findings underscore the critical housekeeping role of Ubr1 and arginylation-dependent endophagy/autophagy during endo-lysosomal proteostasis perturbations and suggest a link of Ubr1 to Ca2+ homeostasis and proteins implicated in various diseases including cancers and brain disorders.


Asunto(s)
Autofagia/fisiología , Calcio/metabolismo , Endosomas/metabolismo , Proteostasis/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Arginina/metabolismo , Células CHO , Línea Celular Tumoral , Cricetulus , Células HeLa , Humanos , Lisosomas/metabolismo , Proteolisis , Transducción de Señal/fisiología , Ubiquitina/metabolismo
2.
Elife ; 102021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34723793

RESUMEN

Absence of the astrocyte-specific membrane protein MLC1 is responsible for megalencephalic leukoencephalopathy with subcortical cysts (MLC), a rare type of leukodystrophy characterized by early-onset macrocephaly and progressive white matter vacuolation that lead to ataxia, spasticity, and cognitive decline. During postnatal development (from P5 to P15 in the mouse), MLC1 forms a membrane complex with GlialCAM (another astrocytic transmembrane protein) at the junctions between perivascular astrocytic processes. Perivascular astrocytic processes along with blood vessels form the gliovascular unit. It was not previously known how MLC1 influences the physiology of the gliovascular unit. Here, using the Mlc1 knock-out mouse model of MLC, we demonstrated that MLC1 controls the postnatal development and organization of perivascular astrocytic processes, vascular smooth muscle cell contractility, neurovascular coupling, and intraparenchymal interstitial fluid clearance. Our data suggest that MLC is a developmental disorder of the gliovascular unit, and perivascular astrocytic processes and vascular smooth muscle cell maturation defects are primary events in the pathogenesis of MLC and therapeutic targets for this disease.


Asunto(s)
Moléculas de Adhesión Celular Neurona-Glia/genética , Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo
3.
Hum Mutat ; 42(10): 1215-1220, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34212451

RESUMEN

De novo rare damaging variants in genes involved in critical developmental pathways, notably regulation of synaptic transmission, have emerged as a frequent cause of neurodevelopmental disorders (NDD). NDD show great locus heterogeneity and for many of the associated genes, there is substantial phenotypic diversity, including epilepsy, intellectual disability, autism spectrum disorder, movement disorders, and combinations thereof. We report two unrelated patients, a young girl with early-onset refractory epilepsy, severe disability, and progressive cerebral and cerebellar atrophy, and a second girl with mild dysmorphism, global developmental delay, and moderate intellectual disability in whom trio-based whole-exome sequencing analysis uncovered de novo missense variants in CHRM1. Biochemical analyses of one of the NDD-associated variants proved that it caused a reduction in protein levels and impaired cellular trafficking. In addition, the mutated receptor showed defective activation of intracellular signaling pathways. Our data strengthen the concept that brain-reduced muscarinic signaling lowers the seizure threshold and severely impairs neurodevelopment.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Epilepsia/genética , Femenino , Humanos , Discapacidad Intelectual/genética , Mutación , Trastornos del Neurodesarrollo/genética , Receptor Muscarínico M1/genética , Receptores Muscarínicos/genética
4.
Neuron ; 109(15): 2427-2442.e10, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34171291

RESUMEN

Astrocytes extensively infiltrate the neuropil to regulate critical aspects of synaptic development and function. This process is regulated by transcellular interactions between astrocytes and neurons via cell adhesion molecules. How astrocytes coordinate developmental processes among one another to parse out the synaptic neuropil and form non-overlapping territories is unknown. Here we identify a molecular mechanism regulating astrocyte-astrocyte interactions during development to coordinate astrocyte morphogenesis and gap junction coupling. We show that hepaCAM, a disease-linked, astrocyte-enriched cell adhesion molecule, regulates astrocyte competition for territory and morphological complexity in the developing mouse cortex. Furthermore, conditional deletion of Hepacam from developing astrocytes significantly impairs gap junction coupling between astrocytes and disrupts the balance between synaptic excitation and inhibition. Mutations in HEPACAM cause megalencephalic leukoencephalopathy with subcortical cysts in humans. Therefore, our findings suggest that disruption of astrocyte self-organization mechanisms could be an underlying cause of neural pathology.


Asunto(s)
Astrocitos/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Animales , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Ratones , Ratas
5.
Hum Mol Genet ; 30(17): 1649-1665, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34100078

RESUMEN

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.


Asunto(s)
Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Receptores Acoplados a Proteínas G/genética , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/genética , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Proteínas de Ciclo Celular/genética , Canales de Cloruro/genética , Quistes/metabolismo , Células HEK293 , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo
6.
Glia ; 69(4): 817-841, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33058289

RESUMEN

Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Encéfalo , Neuroglía , Neuronas
7.
Hum Mol Genet ; 29(7): 1107-1120, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31960914

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a type of leukodystrophy characterized by white matter edema, and it is caused mainly by recessive mutations in MLC1 and GLIALCAM genes. These variants are called MLC1 and MLC2A with both types of patients sharing the same clinical phenotype. In addition, dominant mutations in GLIALCAM have also been identified in a subtype of MLC patients with a remitting phenotype. This variant has been named MLC2B. GLIALCAM encodes for an adhesion protein containing two immunoglobulin (Ig) domains and it is needed for MLC1 targeting to astrocyte-astrocyte junctions. Most mutations identified in GLIALCAM abolish GlialCAM targeting to junctions. However, it is unclear why some mutations behave as recessive or dominant. Here, we used a combination of biochemistry methods with a new developed anti-GlialCAM nanobody, double-mutants and cysteine cross-links experiments, together with computer docking, to create a structural model of GlialCAM homo-interactions. Using this model, we suggest that dominant mutations affect different GlialCAM-GlialCAM interacting surfaces in the first Ig domain, which can occur between GlialCAM molecules present in the same cell (cis) or present in neighbouring cells (trans). Our results provide a framework that can be used to understand the molecular basis of pathogenesis of all identified GLIALCAM mutations.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas de la Membrana/genética , Conformación Proteica , Astrocitos , Encéfalo/patología , Encéfalo/ultraestructura , Proteínas de Ciclo Celular/ultraestructura , Cisteína/genética , Quistes/química , Quistes/patología , Edema/genética , Edema/patología , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Humanos , Proteínas de la Membrana/ultraestructura , Simulación del Acoplamiento Molecular , Mutación , Fenotipo , Multimerización de Proteína , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Sustancia Blanca/ultraestructura
8.
Orphanet J Rare Dis ; 14(1): 268, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31752924

RESUMEN

BACKGROUND: Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a rare type of leukodystrophy characterized by astrocyte and myelin vacuolization, epilepsy and early-onset macrocephaly. MLC is caused by mutations in MLC1 or GLIALCAM, coding for two membrane proteins with an unknown function that form a complex specifically expressed in astrocytes at cell-cell junctions. Recent studies in Mlc1-/- or Glialcam-/- mice and mlc1-/- zebrafish have shown that MLC1 regulates glial surface levels of GlialCAM in vivo and that GlialCAM is also required for MLC1 expression and localization at cell-cell junctions. METHODS: We have generated and analysed glialcama-/- zebrafish. We also generated zebrafish glialcama-/- mlc1-/- and mice double KO for both genes and performed magnetic resonance imaging, histological studies and biochemical analyses. RESULTS: glialcama-/- shows megalencephaly and increased fluid accumulation. In both zebrafish and mice, this phenotype is not aggravated by additional elimination of mlc1. Unlike mice, mlc1 protein expression and localization are unaltered in glialcama-/- zebrafish, possibly because there is an up-regulation of mlc1 mRNA. In line with these results, MLC1 overexpressed in Glialcam-/- mouse primary astrocytes is located at cell-cell junctions. CONCLUSIONS: This work indicates that the two proteins involved in the pathogenesis of MLC, GlialCAM and MLC1, form a functional unit, and thus, that loss-of-function mutations in these genes cause leukodystrophy through a common pathway.


Asunto(s)
Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Proteínas de la Membrana/metabolismo , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Astrocitos/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/genética , Mutación con Pérdida de Función/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Vaina de Mielina/genética , Proteínas del Tejido Nervioso/genética , Pez Cebra , Proteínas de Pez Cebra/metabolismo
9.
Mol Neurobiol ; 56(12): 8237-8254, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31209783

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts protein-1 (MLC1) is a membrane protein expressed by perivascular astrocytes. MLC1 mutations cause MLC, an incurable leukodystrophy characterized by macrocephaly, brain edema, cysts, myelin vacuolation, and astrocytosis, leading to cognitive/motor impairment and epilepsy. Although its function is unknown, MLC1 favors regulatory volume decrease after astrocyte osmotic swelling and down-regulates intracellular signaling pathways controlling astrocyte activation and proliferation. By combining analysis of human brain tissues with in vitro experiments, here we investigated MLC1 role in astrocyte activation during neuroinflammation, a pathological condition exacerbating patient symptoms. MLC1 upregulation was observed in brain tissues from multiple sclerosis, Alzheimer's, and Creutzfeld-Jacob disease, all pathologies characterized by strong astrocytosis and release of inflammatory cytokines, particularly IL-1ß. Using astrocytoma lines overexpressing wild-type (WT) or mutated MLC1 and astrocytes from control and Mlc1 knock-out (KO) mice, we found that IL-1ß stimulated WT-MLC1 plasma membrane expression in astrocytoma cells and control primary astrocytes. In astrocytoma, WT-MLC1 inhibited the activation of IL-1ß-induced inflammatory signals (pERK, pNF-kB) that, conversely, were constitutively activated in mutant expressing cells or abnormally upregulated in KO astrocytes. WT-MLC1+ cells also expressed reduced levels of the astrogliosis marker pSTAT3. We then monitored MLC1 expression timing in a demyelinating/remyelinating murine cerebellar organotypic culture model where, after the demyelination and release of inflammatory cytokines, recovery processes occur, revealing MLC1 upregulation in these latter phases. Altogether, these findings suggest that by modulating specific pathways, MLC1 contributes to restore astrocyte homeostasis after inflammation, providing the opportunity to identify drug target molecules to slow down disease progression.


Asunto(s)
Astrocitos/patología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Transducción de Señal , Adulto , Anciano , Enfermedad de Alzheimer/patología , Animales , Astrocitos/metabolismo , Membrana Celular/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Ratones Noqueados , Persona de Mediana Edad , Modelos Biológicos , Mutación/genética , FN-kappa B/metabolismo , Fosforilación , Ratas , Regulación hacia Arriba
10.
Int J Mol Sci ; 20(5)2019 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-30818802

RESUMEN

Astrocytes are the most abundant cell type in the CNS (central nervous system). They exert multiple functions during development and in the adult CNS that are essential for brain homeostasis. Both cation and anion channel activities have been identified in astrocytes and it is believed that they play key roles in astrocyte function. Whereas the proteins and the physiological roles assigned to cation channels are becoming very clear, the study of astrocytic chloride channels is in its early stages. In recent years, we have moved from the identification of chloride channel activities present in astrocyte primary culture to the identification of the proteins involved in these activities, the determination of their 3D structure and attempts to gain insights about their physiological role. Here, we review the recent findings related to the main chloride channels identified in astrocytes: the voltage-dependent ClC-2, the calcium-activated bestrophin, the volume-activated VRAC (volume-regulated anion channel) and the stress-activated Maxi-Cl-. We discuss key aspects of channel biophysics and structure with a focus on their role in glial physiology and human disease.


Asunto(s)
Astrocitos/metabolismo , Encefalopatías/metabolismo , Encefalopatías/patología , Encéfalo/metabolismo , Encéfalo/patología , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Homeostasis , Animales , Humanos , Modelos Biológicos
11.
Neurobiol Dis ; 119: 88-99, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076890

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/metabolismo , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Astrocitos/química , Astrocitos/patología , Proteínas de Ciclo Celular , Células Cultivadas , Quistes/patología , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Proteínas/análisis , Proteínas/genética , Ratas , Xenopus
12.
Physiol Rep ; 6(2)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29380539

RESUMEN

Modulation of CaV 2.1 channel activity plays a key role in interneuronal communication and synaptic plasticity. SNAREs interact with a specific synprint site at the second intracellular loop (LII-III) of the CaV 2.1 pore-forming α1A subunit to optimize neurotransmitter release from presynaptic terminals by allowing secretory vesicles docking near the Ca2+ entry pathway, and by modulating the voltage dependence of channel steady-state inactivation. Ca2+ influx through CaV 2.1 also promotes channel inactivation. This process seems to involve Ca2+ -calmodulin interaction with two adjacent sites in the α1A carboxyl tail (C-tail) (the IQ-like motif and the Calmodulin-Binding Domain (CBD) site), and contributes to long-term potentiation and spatial learning and memory. Besides, binding of regulatory ß subunits to the α interaction domain (AID) at the first intracellular loop (LI-II) of α1A determines the degree of channel inactivation by both voltage and Ca2+ . Here, we explore the cross talk between ß subunits, Ca2+ , and syntaxin-1A-modulated CaV 2.1 inactivation, highlighting the α1A domains involved in such process. ß3 -containing CaV 2.1 channels show syntaxin-1A-modulated but no Ca2+ -dependent steady-state inactivation. Conversely, ß2a -containing CaV 2.1 channels show Ca2+ -dependent but not syntaxin-1A-modulated steady-state inactivation. A LI-II deletion confers Ca2+ -dependent inactivation and prevents modulation by syntaxin-1A in ß3 -containing CaV 2.1 channels. Mutation of the IQ-like motif, unlike CBD deletion, abolishes Ca2+ -dependent inactivation and confers modulation by syntaxin-1A in ß2a -containing CaV 2.1 channels. Altogether, these results suggest that LI-II structural modifications determine the regulation of CaV 2.1 steady-state inactivation either by Ca2+ or by SNAREs but not by both.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Señalización del Calcio/fisiología , Proteínas SNARE/metabolismo , Células HEK293 , Humanos , Receptor Cross-Talk/fisiología , Transmisión Sináptica/fisiología , Sintaxina 1/metabolismo
13.
Eur J Med Genet ; 61(1): 50-60, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29079544

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy characterized by dysfunction of the role of glial cells in controlling brain fluid and ion homeostasis. Patients affected by MLC present macrocephaly, cysts and white matter vacuolation, which lead to motor and cognitive impairments. To date, there is no treatment for MLC, only supportive care. MLC is caused by mutations in the MLC1 and GLIALCAM genes. MLC1 is a membrane protein with low identity to the Kv1.1 potassium channel and GlialCAM belongs to an adhesion molecule family. Both proteins form a complex with an as-yet-unknown function that is expressed mainly in the astrocytes surrounding the blood-brain barrier and in Bergmann glia. GlialCAM also acts as an auxiliary subunit of the chloride channel ClC-2, thus regulating its localization at cell-cell junctions and modifying its functional properties by affecting the common gate of ClC-2. Recent studies in Mlc1-, GlialCAM- and Clcn2-knockout mice or Mlc1-knockout zebrafish have provided fresh insight into the pathophysiology of MLC and further details about the molecular interactions between these three proteins. Additional studies have shown that GlialCAM/MLC1 also regulates other ion channels (TRPV4, VRAC) or transporters (Na+/K+-ATPase) in a not-understood manner. Furthermore, it has been shown that GlialCAM/MLC1 may influence signal transduction mechanisms, thereby affecting other proteins not related with transport such as the EGF receptor. Here, we offer a personal biochemical retrospective of the work that has been performed to gain knowledge of the pathophysiology of MLC, and we discuss future strategies that may be used to identify therapeutic solutions for MLC patients.


Asunto(s)
Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas/genética , Animales , Encéfalo/metabolismo , Proteínas de Ciclo Celular , Quistes/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica , Proteínas/química , Proteínas/metabolismo
14.
J Physiol ; 595(22): 6993-7008, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28905383

RESUMEN

KEY POINTS: Characterisation of most mutations found in CLCN2 in patients with CC2L leukodystrophy show that they cause a reduction in function of the chloride channel ClC-2. GlialCAM, a regulatory subunit of ClC-2 in glial cells and involved in the leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC), increases the activity of a ClC-2 mutant by affecting ClC-2 gating and by stabilising the mutant at the plasma membrane. The stabilisation of ClC-2 at the plasma membrane by GlialCAM depends on its localisation at cell-cell junctions. The membrane protein MLC1, which is defective in MLC, also contributes to the stabilisation of ClC-2 at the plasma membrane, providing further support for the view that GlialCAM, MLC1 and ClC-2 form a protein complex in glial cells. ABSTRACT: Mutations in CLCN2 have been recently identified in patients suffering from a type of leukoencephalopathy involving intramyelinic oedema. Here, we characterised most of these mutations that reduce the function of the chloride channel ClC-2 and impair its plasma membrane (PM) expression. Detailed biochemical and electrophysiological analyses of the Ala500Val mutation revealed that defective gating and increased cellular and PM turnover contributed to defective A500V-ClC-2 functional expression. Co-expression of the adhesion molecule GlialCAM, which forms a tertiary complex with ClC-2 and megalencephalic leukoencephalopathy with subcortical cysts 1 (MLC1), rescued the functional expression of the mutant by modifying its gating properties. GlialCAM also restored the PM levels of the channel by impeding its turnover at the PM. This rescue required ClC-2 localisation to cell-cell junctions, since a GlialCAM mutant with compromised junctional localisation failed to rescue the impaired stability of mutant ClC-2 at the PM. Wild-type, but not mutant, ClC-2 was also stabilised by MLC1 overexpression. We suggest that leukodystrophy-causing CLCN2 mutations reduce the functional expression of ClC-2, which is partly counteracted by GlialCAM/MLC1-mediated increase in the gating and stability of the channel.


Asunto(s)
Canales de Cloruro/metabolismo , Activación del Canal Iónico , Leucoencefalopatías/genética , Mutación , Animales , Canales de Cloruro CLC-2 , Membrana Celular/metabolismo , Células Cultivadas , Canales de Cloruro/genética , Cloruros/metabolismo , Células HEK293 , Células HeLa , Humanos , Neuroglía/metabolismo , Estabilidad Proteica , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Uniones Estrechas/metabolismo , Xenopus
15.
Hum Mol Genet ; 26(13): 2436-2450, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28398517

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Encefalopatías/patología , Canales de Cloruro CLC-2 , Canales de Calcio Tipo L/genética , Canales de Cloruro , Quistes/genética , Células HEK293 , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Transporte de Proteínas/genética
16.
J Physiol ; 593(18): 4165-80, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26033718

RESUMEN

KEY POINTS: The extracellular domain of GlialCAM is necessary for its targeting to cell junctions, as well as for interactions with itself and MLC1 and ClC-2. The C-terminus of GlialCAM is not necessary for interaction but is required for targeting to cell junctions. The first three residues of the transmembrane segment of GlialCAM are required for GlialCAM-mediated ClC-2 activation. ABSTRACT: Mutations in the genes encoding the astrocytic protein MLC1, the cell adhesion molecule GlialCAM or the Cl(-) channel ClC-2 underlie human leukodystrophies. GlialCAM binds to itself, to MLC1 and to ClC-2, and directs these proteins to cell-cell contacts. In addition, GlialCAM dramatically activates ClC-2 mediated currents. In the present study, we used mutagenesis studies combined with functional and biochemical analyses to determine which parts of GlialCAM are required to perform these cellular functions. We found that the extracellular domain of GlialCAM is necessary for cell junction targeting and for mediating interactions with itself or with MLC1 and ClC-2. The C-terminus is also necessary for proper targeting to cell-cell junctions but is not required for the biochemical interaction. Finally, we identified the first three amino acids of the transmembrane segment of GlialCAM as being essential for the activation of ClC-2 currents but not for targeting or biochemical interaction. Our results provide new mechanistic insights concerning the regulation of the cell biology and function of MLC1 and ClC-2 by GlialCAM.


Asunto(s)
Encefalopatías/metabolismo , Canales de Cloruro/metabolismo , Proteínas de la Membrana/metabolismo , Subunidades de Proteína/metabolismo , Proteínas/metabolismo , Astrocitos/metabolismo , Encefalopatías/genética , Canales de Cloruro CLC-2 , Proteínas de Ciclo Celular , Línea Celular , Línea Celular Tumoral , Canales de Cloruro/genética , Células HEK293 , Células HeLa , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/genética , Mutación/genética , Subunidades de Proteína/genética , Transporte de Proteínas/genética , Proteínas/genética
17.
Biophys J ; 107(5): 1105-1116, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25185546

RESUMEN

GlialCAM, a glial cell adhesion molecule mutated in megalencephalic leukoencephalopathy with subcortical cysts, targets the CLC-2 Cl(-) channel to cell contacts in glia and activates CLC-2 currents in vitro and in vivo. We found that GlialCAM clusters all CLC channels at cell contacts in vitro and thus studied GlialCAM interaction with CLC channels to investigate the mechanism of functional activation. GlialCAM slowed deactivation kinetics of CLC-Ka/barttin channels and increased CLC-0 currents opening the common gate and slowing its deactivation. No functional effect was seen for common gate deficient CLC-0 mutants. Similarly, GlialCAM targets the common gate deficient CLC-2 mutant E211V/H816A to cell contacts, without altering its function. Thus, GlialCAM is able to interact with all CLC channels tested, targeting them to cell junctions and activating them by stabilizing the open configuration of the common gate. These results are important to better understand the physiological role of GlialCAM/CLC-2 interaction.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas/metabolismo , Animales , Proteínas de Ciclo Celular , Canales de Cloruro/genética , Células HEK293 , Humanos , Iones/química , Cinética , Potenciales de la Membrana/fisiología , Mutación , Oocitos , Ratas , Temperatura , Torpedo , Transfección , Xenopus laevis , Zinc/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA