Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 37(21): 5309-5318, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28450545

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal-recessive disorder characterized by severe, often fatal muscle weakness due to loss of motor neurons. SMA patients have deletions and other mutations of the survival of motor neuron 1 (SMN1) gene, resulting in decreased SMN protein. Astrocytes are the primary support cells of the CNS and are responsible for glutamate clearance, metabolic support, response to injury, and regulation of signal transmission. Astrocytes have been implicated in SMA as in in other neurodegenerative disorders. Astrocyte-specific rescue of SMN protein levels has been shown to mitigate disease manifestations in mice. However, the mechanism by which SMN deficiency in astrocytes may contribute to SMA is unclear and what aspect of astrocyte activity is lacking is unknown. Therefore, it is worthwhile to identify defects in SMN-deficient astrocytes that compromise normal function. We show here that SMA astrocyte cultures derived from mouse spinal cord of both sexes are deficient in supporting both WT and SMN-deficient motor neurons derived from male, female, and mixed-sex sources and that this deficiency may be mitigated with secreted factors. In particular, SMN-deficient astrocytes have decreased levels of monocyte chemoactive protein 1 (MCP1) secretion compared with controls and MCP1 restoration stimulates outgrowth of neurites from cultured motor neurons. Correction of MCP1 deficiency may thus be a new therapeutic approach to SMA.SIGNIFICANCE STATEMENT Spinal muscular atrophy (SMA) is caused by the loss of motor neurons, but astrocyte dysfunction also contributes to the disease in mouse models. Monocyte chemoactive protein 1 (MCP1) has been shown to be neuroprotective and is released by astrocytes. Here, we report that MCP1 levels are decreased in SMA mice and that replacement of deficient MCP1 increases differentiation and neurite length of WT and SMN-deficient motor-neuron-like cells in cell culture. This study reveals a novel aspect of astrocyte dysfunction in SMA and indicates a possible approach for improving motor neuron growth and survival in this disease.


Subject(s)
Astrocytes/metabolism , Chemokine CCL2/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/genetics , Animals , Astrocytes/cytology , Cells, Cultured , Chemokine CCL2/genetics , Female , Humans , Male , Mice , Motor Neurons/cytology , Spinal Cord/cytology , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/metabolism
2.
Exp Neurol ; 292: 145-153, 2017 06.
Article in English | MEDLINE | ID: mdl-28342750

ABSTRACT

Downregulation in the astroglial glutamate transporter EAAT2 in amyotrophic lateral sclerosis (ALS) patients and mutant SOD1 mouse models of ALS is believed to contribute to the death of motor neurons by excitotoxicity. We previously reported that caspase-3 cleaves EAAT2 at a unique cleavage consensus site located in its c-terminus domain, a proteolytic cleavage that also occurs in vivo in the mutant SOD1 mouse model of ALS and leads to accumulation of a sumoylated EAAT2 C-terminus fragment (CTE-SUMO1) beginning around onset of disease. CTE-SUMO1 accumulates in PML nuclear bodies of astrocytes and causes them to alter their mature phenotypes and secrete factors toxic to motor neurons. Here, we report that mutating the caspase-3 consensus site in the EAAT2 sequence with an aspartate to asparagine mutation (D504N), thereby inhibiting caspase-3 cleavage of EAAT2, confers protection to the SOD1-G93A mouse. EAAT2-D504N knock-in mutant mice were generated and crossed with SOD1-G93A mice to assess the in vivo pathogenic relevance for ALS symptoms of EAAT2 cleavage. The mutation did not affect normal EAAT2 function nor non-ALS mice. In agreement with the timing of CTE-SUMO1 accumulation, while onset of disease was not affected, the mutation caused an extension in progression time, a delay in the development of hindlimb and forelimb muscle weakness, and a significant increase in the lifespan of SOD1-G93A mice.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Caspase 3/metabolism , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/metabolism , Mutation/genetics , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Disease Models, Animal , Disease Progression , Glutamic Acid/genetics , Intranuclear Inclusion Bodies/metabolism , Mice , Motor Neurons/metabolism , Motor Neurons/pathology
3.
Neurobiol Dis ; 88: 118-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792401

ABSTRACT

The development of therapeutics for neurological disorders is constrained by limited access to the central nervous system (CNS). ATP-binding cassette (ABC) transporters, particularly P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), are expressed on the luminal surface of capillaries in the CNS and transport drugs out of the endothelium back into the blood against the concentration gradient. Survival motor neuron (SMN) protein, which is deficient in spinal muscular atrophy (SMA), is a target of the ubiquitin proteasome system. Inhibiting the proteasome in a rodent model of SMA with bortezomib increases SMN protein levels in peripheral tissues but not the CNS, because bortezomib has poor CNS penetrance. We sought to determine if we could inhibit SMN degradation in the CNS of SMA mice with a combination of bortezomib and the ABC transporter inhibitor tariquidar. In cultured cells we show that bortezomib is a substrate of P-gp. Mass spectrometry analysis demonstrated that intraperitoneal co-administration of tariquidar increased the CNS penetrance of bortezomib, and reduced proteasome activity in the brain and spinal cord. This correlated with increased SMN protein levels and improved survival and motor function of SMA mice. These findings show that CNS penetrance of treatment for this neurological disorder can be improved by inhibiting drug efflux at the blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/metabolism , Bortezomib/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Age Factors , Animals , Animals, Newborn , Antineoplastic Agents/pharmacology , Central Nervous System/cytology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Transgenic , Motor Neurons/drug effects , Proteasome Endopeptidase Complex , Quinolines/pharmacology , Quinolines/therapeutic use , Time Factors , Transfection
4.
Neuromolecular Med ; 15(4): 760-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24062161

ABSTRACT

Emerging lines of evidence suggest a relationship between amyotrophic lateral sclerosis (ALS) and protein sumoylation. Multiple studies have demonstrated that several of the proteins involved in the pathogenesis of ALS, including superoxide dismutase 1, fused in liposarcoma, and TAR DNA-binding protein 43 (TDP-43), are substrates for sumoylation. Additionally, recent studies in cellular and animal models of ALS revealed that sumoylation of these proteins impact their localization, longevity, and how they functionally perform in disease, providing novel areas for mechanistic investigations and therapeutics. In this article, we summarize the current literature examining the impact of sumoylation of critical proteins involved in ALS and discuss the potential impact for the pathogenesis of the disease. In addition, we report and discuss the implications of new evidence demonstrating that sumoylation of a fragment derived from the proteolytic cleavage of the astroglial glutamate transporter, EAAT2, plays a direct role in downregulating the expression levels of full-length EAAT2 by binding to a regulatory region of its promoter.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Nerve Tissue Proteins/physiology , Small Ubiquitin-Related Modifier Proteins/physiology , Sumoylation/physiology , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/metabolism , Calcium Signaling , DNA-Binding Proteins/metabolism , Disease Models, Animal , Excitatory Amino Acid Transporter 2 , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Motor Neurons/metabolism , Muscular Disorders, Atrophic/metabolism , Protein Conformation , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
5.
J Biol Chem ; 287(49): 41139-51, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23060447

ABSTRACT

Mutations in the P4-ATPase ATP8B1 cause the inherited liver disease progressive familial intrahepatic cholestasis. Several of these mutations are located in conserved regions of the transmembrane domain associated with substrate binding and transport. Assays for P4-ATPase-mediated transport in living yeast cells were developed and used to characterize the specificity and kinetic parameters of this transport. Progressive familial intrahepatic cholestasis mutations were introduced into the yeast plasma membrane P4-ATPase Dnf2p, and the effect of these mutations on its catalysis of phospholipid transport were determined. The results of these measurements have implications for the basis of the disease and for the mechanism of phospholipid transit through the enzyme during the reaction cycle.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Cholestasis, Intrahepatic/genetics , Mutation , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Humans , Kinetics , Molecular Sequence Data , Phenotype , Phospholipids/chemistry , Regression Analysis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
6.
Glia ; 59(11): 1719-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21769946

ABSTRACT

Dysregulation of glutamate handling ensuing downregulation of expression and activity levels of the astroglial glutamate transporter EAAT2 is implicated in excitotoxic degeneration of motor neurons in amyotrophic lateral sclerosis (ALS). We previously reported that EAAT2 (a.k.a. GLT-1) is cleaved by caspase-3 at its cytosolic carboxy-terminus domain. This cleavage results in impaired glutamate transport activity and generates a proteolytic fragment (CTE) that we found to be post-translationally conjugated by SUMO1. We show here that this sumoylated CTE fragment accumulates in the nucleus of spinal cord astrocytes of the SOD1-G93A mouse model of ALS at symptomatic stages of disease. Astrocytic expression of CTE, artificially tagged with SUMO1 (CTE-SUMO1) to mimic the native sumoylated fragment, recapitulates the nuclear accumulation pattern of the endogenous EAAT2-derived proteolytic fragment. Moreover, in a co-culture binary system, expression of CTE-SUMO1 in spinal cord astrocytes initiates extrinsic toxicity by inducing caspase-3 activation in motor neuron-derived NSC-34 cells or axonal growth impairment in primary motor neurons. Interestingly, prolonged nuclear accumulation of CTE-SUMO1 is intrinsically toxic to spinal cord astrocytes, although this gliotoxic effect of CTE-SUMO1 occurs later than the indirect, noncell autonomous toxic effect on motor neurons. As more evidence on the implication of SUMO substrates in neurodegenerative diseases emerges, our observations strongly suggest that the nuclear accumulation in spinal cord astrocytes of a sumoylated proteolytic fragment of the astroglial glutamate transporter EAAT2 could participate to the pathogenesis of ALS and suggest a novel, unconventional role for EAAT2 in motor neuron degeneration.


Subject(s)
Excitatory Amino Acid Transporter 2/toxicity , Motor Neurons/drug effects , SUMO-1 Protein/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/metabolism , Axons/physiology , Axons/ultrastructure , Caspase 3/metabolism , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Coculture Techniques , Excitatory Amino Acid Transporter 2/chemistry , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Transgenic , Microarray Analysis , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Real-Time Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase-1
7.
Antioxid Redox Signal ; 11(7): 1587-602, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19413484

ABSTRACT

Responsible for the majority of excitatory activity in the central nervous system (CNS), glutamate interacts with a range of specific receptor and transporter systems to establish a functional synapse. Excessive stimulation of glutamate receptors causes excitotoxicity, a phenomenon implicated in both acute and chronic neurodegenerative diseases [e.g., ischemia, Huntington's disease, and amyotrophic lateral sclerosis (ALS)]. In physiology, excitotoxicity is prevented by rapid binding and clearance of synaptic released glutamate by high-affinity, Na(+)-dependent glutamate transporters and amplified by defects to the glutamate transporter and receptor systems. ALS pathogenetic mechanisms are not completely understood and characterized, but excitotoxicity has been regarded as one firm mechanism implicated in the disease because of data obtained from ALS patients and animal and cellular models as well as inferred by the documented efficacy of riluzole, a generic antiglutamatergic drug, has in patients. In this article, we critically review the several lines of evidence supporting a role for glutamate-mediated excitotoxicity in the death of motor neurons occurring in ALS, putting a particular emphasis on the impairment of the glutamate-transport system.


Subject(s)
Amino Acid Transport System X-AG/physiology , Amyotrophic Lateral Sclerosis/physiopathology , Motor Neurons , Animals , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...