Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Hum Mol Genet ; 32(23): 3263-3275, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37658769

ABSTRACT

The COPI coatomer subunit α-COP has been shown to co-precipitate mRNA in multiple settings, but it was unclear whether the interaction with mRNA was direct or mediated by interaction with an adapter protein. The COPI complex often interacts with proteins via C-terminal dilysine domains. A search for candidate RNA binding proteins with C-terminal dilysine motifs yielded Nucleolin, which terminates in a KKxKxx sequence. This protein was an especially intriguing candidate as it has been identified as an interacting partner for Survival Motor Neuron protein (SMN). Loss of SMN causes the neurodegenerative disease Spinal Muscular Atrophy. We have previously shown that SMN and α-COP interact and co-migrate in axons, and that overexpression of α-COP reduced phenotypic severity in cell culture and animal models of SMA. We show here that in an mRNA independent manner, endogenous Nucleolin co-precipitates endogenous α-COP and ε-COP but not ß-COP which may reflect an interaction with the so-called B-subcomplex rather a complete COPI heptamer. The ability of Nucleolin to bind to α-COP requires the presence of the C-terminal KKxKxx domain of Nucleolin. Furthermore, we have generated a point mutant in the WD40 domain of α-COP which eliminates its ability to co-precipitate Nucleolin but does not interfere with precipitation of partners mediated by non-KKxKxx motifs such as the kainate receptor subunit 2. We propose that via interaction between the C-terminal dilysine motif of Nucleolin and the WD40 domain of α-COP, Nucleolin acts an adaptor to allow α-COP to interact with a population of mRNA.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Animals , Coatomer Protein/genetics , Protein Binding , Phosphoproteins/genetics , Phosphoproteins/metabolism , Muscular Atrophy, Spinal/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleolin
2.
Biochem Biophys Res Commun ; 514(2): 530-537, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31060774

ABSTRACT

We report that expression of the α-COP protein rescues disease phenotype in a severe mouse model of Spinal Muscular Atrophy (SMA). Lentiviral particles expressing α-COP were injected directly into the testes of genetically pure mouse strain of interest resulting in infection of the spermatagonial stem cells. α-COP was stably expressed in brain, skeletal muscle, and spinal cord without altering SMN protein levels. SMA mice transgenic for α-COP live significantly longer than their non-transgenic littermates, and showed increased body mass and normal muscle morphology at postnatal day 15. We previously reported that binding between SMN and α-COP is required for restoration of neurite outgrowth in cells lacking SMN, and we report similar finding here. Lentiviral-mediated transgenic expression of SMN where the dilysine domain in exon 2b was mutated was not able to rescue the SMA phenotype despite robust expression of the mutant SMN protein in brain, muscle and spinal cord. These results demonstrate that α-COP is a validated modifier of SMA disease phenotype in a mammalian, vertebrate model and is a potential target for development of future SMN-independent therapeutic interventions.


Subject(s)
Coatomer Protein/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Coatomer Protein/metabolism , Disease Models, Animal , Exons , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Mutation , Phenotype , Protein Binding , Signal Transduction , Spinal Cord/pathology , Survival Analysis , Survival of Motor Neuron 1 Protein/metabolism
3.
Neurobiol Aging ; 101: 57-69, 2021 05.
Article in English | MEDLINE | ID: mdl-33582567

ABSTRACT

Understanding the cellular processes that lead to Alzheimer's disease (AD) is critical, and one key lies in the genetics of families with histories of AD. Mutations a complex known as COPI were found in families with AD. The COPI complex is involved in protein processing and trafficking. Intriguingly, several recent publications have found components of the COPI complex can affect the metabolism of pathogenic AD proteins. We reduced levels of the COPI subunit α-COP, altering maturation and cleavage of amyloid precursor protein (APP), resulting in decreased release of Aß-42 and decreased accumulation of the AICD. Depletion of α-COP reduced uptake of proteopathic Tau seeds and reduces intracellular Tau self-association. Expression of AD-associated mutant α-COP altered APP processing, resulting in increased release of Aß-42 and increased intracellular Tau aggregation and release of Tau oligomers. These results show that COPI coatomer function modulates processing of both APP and Tau, and expression of AD-associated α-COP confers a toxic gain of function, resulting in potentially pathogenic changes in both APP and Tau.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Coat Protein Complex I/genetics , Coat Protein Complex I/physiology , Coatomer Protein/genetics , Coatomer Protein/physiology , Mutation/genetics , Mutation/physiology , Peptide Fragments/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Cells, Cultured , Mice
4.
PLoS One ; 11(10): e0163954, 2016.
Article in English | MEDLINE | ID: mdl-27736905

ABSTRACT

Spinal muscular atrophy (SMA) is an intractable neurodegenerative disease afflicting 1 in 6-10,000 live births. One of the key functions of the SMN protein is regulation of spliceosome assembly. Reduced levels of the SMN protein that are observed in SMA have been shown to result in aberrant mRNA splicing. SMN-dependent mis-spliced transcripts in motor neurons may cause stresses that are particularly harmful and may serve as potential targets for the treatment of motor neuron disease or as biomarkers in the SMA patient population. We performed deep RNA sequencing using motor neuron-like NSC-34 cells to screen for SMN-dependent mRNA processing changes that occur following acute depletion of SMN. We identified SMN-dependent splicing changes, including an intron retention event that results in the production of a truncated Rit1 transcript. This intron-retained transcript is stable and is mis-spliced in spinal cord from symptomatic SMA mice. Constitutively active Rit1 ameliorated the neurite outgrowth defect in SMN depleted NSC-34 cells, while expression of the truncated protein product of the mis-spliced Rit1 transcript inhibited neurite extension. These results reveal new insights into the biological consequence of SMN-dependent splicing in motor neuron-like cells.


Subject(s)
Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , RNA Splicing , RNA, Messenger/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Animals , Cell Line , Disease Models, Animal , Exons , Female , Gene Expression Regulation , Introns , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL