Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 298(10): 102423, 2022 10.
Article in English | MEDLINE | ID: mdl-36030824

ABSTRACT

Gamma-aminobutyric acid type A (GABAA) receptors are the primary inhibitory neurotransmitter-gated ion channels in the mammalian central nervous system. Maintenance of GABAA receptor protein homeostasis (proteostasis) in cells utilizing its interacting proteins is essential for the function of GABAA receptors. However, how the proteostasis network orchestrates GABAA receptor biogenesis in the endoplasmic reticulum is not well understood. Here, we employed a proteomics-based approach to systematically identify the interactomes of GABAA receptors. We carried out a quantitative immunoprecipitation-tandem mass spectrometry analysis utilizing stable isotope labeling by amino acids in cell culture. Furthermore, we performed comparative proteomics by using both WT α1 subunit and a misfolding-prone α1 subunit carrying the A322D variant as the bait proteins. We identified 125 interactors for WT α1-containing receptors, 105 proteins for α1(A322D)-containing receptors, and 54 overlapping proteins within these two interactomes. Our bioinformatics analysis identified potential GABAA receptor proteostasis network components, including chaperones, folding enzymes, trafficking factors, and degradation factors, and we assembled a model of their potential involvement in the cellular folding, degradation, and trafficking pathways for GABAA receptors. In addition, we verified endogenous interactions between α1 subunits and selected interactors by using coimmunoprecipitation in mouse brain homogenates. Moreover, we showed that TRIM21 (tripartite motif containing-21), an E3 ubiquitin ligase, positively regulated the degradation of misfolding-prone α1(A322D) subunits selectively. This study paves the way for understanding the molecular mechanisms as well as fine-tuning of GABAA receptor proteostasis to ameliorate related neurological diseases such as epilepsy.


Subject(s)
Proteostasis , Receptors, GABA-A , Animals , Mice , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , gamma-Aminobutyric Acid/metabolism , Proteomics , Receptors, GABA-A/metabolism
2.
Cell Chem Biol ; 28(1): 46-59.e7, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32888501

ABSTRACT

Proteostasis deficiency in mutated ion channels leads to a variety of ion channel diseases that are caused by excessive endoplasmic reticulum-associated degradation (ERAD) and inefficient membrane trafficking. We investigated proteostasis maintenance of γ-aminobutyric acid type A (GABAA) receptors, the primary mediators of neuronal inhibition in the mammalian central nervous system. We screened a structurally diverse, Food and Drug Administration-approved drug library and identified dinoprost (DNP) and dihydroergocristine (DHEC) as highly efficacious enhancers of surface expression of four epilepsy-causing trafficking-deficient mutant receptors. Furthermore, DNP and DHEC restore whole-cell and synaptic currents by incorporating mutated subunits into functional receptors. Mechanistic studies revealed that both drugs reduce subunit degradation by attenuating the Grp94/Hrd1/Sel1L/VCP-mediated ERAD pathway and enhance the subunit folding by promoting subunit interactions with major GABAA receptors-interacting chaperones, BiP and calnexin. In summary, we report that DNP and DHEC remodel the endoplasmic reticulum proteostasis network to restore the functional surface expression of mutant GABAA receptors.


Subject(s)
Dihydroergocristine/pharmacology , Dinoprost/pharmacology , Epilepsy/drug therapy , Proteostasis/drug effects , Receptors, GABA-A/metabolism , Cell Line , Endoplasmic Reticulum-Associated Degradation/drug effects , Epilepsy/metabolism , Female , Humans , Male , Receptors, GABA-A/genetics
3.
PLoS One ; 13(11): e0207948, 2018.
Article in English | MEDLINE | ID: mdl-30481215

ABSTRACT

Biogenesis of membrane proteins is controlled by the protein homeostasis (proteostasis) network. We have been focusing on protein quality control of γ-aminobutyric acid type A (GABAA) receptors, the major inhibitory neurotransmitter-gated ion channels in mammalian central nervous system. Proteostasis deficiency in GABAA receptors causes loss of their surface expression and thus function on the plasma membrane, leading to epilepsy and other neurological diseases. One well-characterized example is the A322D mutation in the α1 subunit that causes its extensive misfolding and expedited degradation in the endoplasmic reticulum (ER), resulting in autosomal dominant juvenile myoclonic epilepsy. We aimed to correct misfolding of the α1(A322D) subunits in the ER as an approach to restore their functional surface expression. Here, we showed that application of BIX, a specific, potent ER resident HSP70 family protein BiP activator, significantly increases the surface expression of the mutant receptors in human HEK293T cells and neuronal SH-SY5Y cells. BIX attenuates the degradation of α1(A322D) and enhances their forward trafficking and function. Furthermore, because BiP is one major target of the two unfolded protein response (UPR) pathways: ATF6 and IRE1, we continued to demonstrate that modest activations of the ATF6 pathway and IRE1 pathway genetically enhance the plasma membrane trafficking of the α1(A322D) protein in HEK293T cells. Our results underlie the potential of regulating the ER proteostasis network to correct loss-of-function protein conformational diseases.


Subject(s)
Endoplasmic Reticulum/metabolism , Proteostasis , Receptors, GABA-A/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Mutation , Neurons/drug effects , Neurons/metabolism , Protein Conformation , Protein Transport/drug effects , Proteostasis/drug effects , Receptors, GABA-A/genetics , Thiocyanates/pharmacology , Unfolded Protein Response/drug effects
4.
Cell Rep ; 21(10): 2895-2910, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212034

ABSTRACT

GADD34, a stress-induced regulatory subunit of the phosphatase PP1, is known to function in hyperosmotic stress through its well-known role in the integrated stress response (ISR) pathway. Adaptation to hyperosmotic stress is important for the health of corneal epithelial cells exposed to changes in extracellular osmolarity, with maladaptation leading to dry eye syndrome. This adaptation includes induction of SNAT2, an endoplasmic reticulum (ER)-Golgi-processed protein, which helps to reverse the stress-induced loss of cell volume and promote homeostasis through amino acid uptake. Here, we show that GADD34 promotes the processing of proteins synthesized on the ER during hyperosmotic stress independent of its action in the ISR. We show that GADD34/PP1 phosphatase activity reverses hyperosmotic-stress-induced Golgi fragmentation and is important for cis- to trans-Golgi trafficking of SNAT2, thereby promoting SNAT2 plasma membrane localization and function. These results suggest that GADD34 is a protective molecule for ocular diseases such as dry eye syndrome.


Subject(s)
Amino Acid Transport System A/metabolism , Protein Phosphatase 1/metabolism , Amino Acid Transport System A/genetics , Amino Acids/metabolism , Blotting, Western , Humans , Osmosis/physiology , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction
5.
J Biol Chem ; 291(18): 9526-39, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26945068

ABSTRACT

Proteostasis maintenance of γ-aminobutyric acid type A (GABAA) receptors dictates their function in controlling neuronal inhibition in mammalian central nervous systems. However, as a multisubunit, multispan, integral membrane protein, even wild type subunits of GABAA receptors fold and assemble inefficiently in the endoplasmic reticulum (ER). Unassembled and misfolded subunits undergo ER-associated degradation (ERAD), but this degradation process remains poorly understood for GABAA receptors. Here, using the α1 subunits of GABAA receptors as a model substrate, we demonstrated that Grp94, a metazoan-specific Hsp90 in the ER lumen, uses its middle domain to interact with the α1 subunits and positively regulates their ERAD. OS-9, an ER-resident lectin, acts downstream of Grp94 to further recognize misfolded α1 subunits in a glycan-dependent manner. This delivers misfolded α1 subunits to the Hrd1-mediated ubiquitination and the valosin-containing protein-mediated extraction pathway. Repressing the initial ERAD recognition step by inhibiting Grp94 enhances the functional surface expression of misfolding-prone α1(A322D) subunits, which causes autosomal dominant juvenile myoclonic epilepsy. This study clarifies a Grp94-mediated ERAD pathway for GABAA receptors, which provides a novel way to finely tune their function in physiological and pathophysiological conditions.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/metabolism , Proteolysis , Receptors, GABA-A/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Substitution , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Mutation, Missense , Receptors, GABA-A/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology
6.
J Biol Chem ; 290(1): 325-37, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25406314

ABSTRACT

GABAA receptors are the primary inhibitory ion channels in the mammalian central nervous system. The A322D mutation in the α1 subunit results in its excessive endoplasmic reticulum-associated degradation at the expense of plasma membrane trafficking, leading to autosomal dominant juvenile myoclonic epilepsy. Presumably, valosin-containing protein (VCP)/p97 extracts misfolded subunits from the endoplasmic reticulum membrane to the cytosolic proteasome for degradation. Here we showed that inhibiting VCP using Eeyarestatin I reduces the endoplasmic reticulum-associated degradation of the α1(A322D) subunit without an apparent effect on its dynamin-1 dependent endocytosis and that this treatment enhances its trafficking. Furthermore, coapplication of Eeyarestatin I and suberanilohydroxamic acid, a known small molecule that promotes chaperone-assisted folding, yields an additive restoration of surface expression of α1(A322D) subunits in HEK293 cells and neuronal SH-SY5Y cells. Consequently, this combination significantly increases GABA-induced chloride currents in whole-cell patch clamping experiments than either chemical compound alone in HEK293 cells. Our findings suggest that VCP inhibition without stress induction, together with folding enhancement, represents a new strategy to restore proteostasis of misfolding-prone GABAA receptors and, therefore, a potential remedy for idiopathic epilepsy.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Endoplasmic Reticulum-Associated Degradation/drug effects , Hydrazones/pharmacology , Hydroxamic Acids/pharmacology , Hydroxyurea/analogs & derivatives , Receptors, GABA-A/chemistry , Action Potentials/drug effects , Action Potentials/physiology , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Adolescent , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chlorides/metabolism , Drug Synergism , Dynamin I/genetics , Dynamin I/metabolism , Endocytosis/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation/genetics , HEK293 Cells , Humans , Hydroxyurea/pharmacology , Myoclonic Epilepsy, Juvenile/genetics , Myoclonic Epilepsy, Juvenile/metabolism , Myoclonic Epilepsy, Juvenile/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Patch-Clamp Techniques , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Protein Folding/drug effects , Protein Stability/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Signal Transduction , Valosin Containing Protein , Vorinostat , gamma-Aminobutyric Acid/metabolism
7.
Pharmacol Res ; 83: 3-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24747662

ABSTRACT

Normal organismal physiology depends on the maintenance of proteostasis in each cellular compartment to achieve a delicate balance between protein synthesis, folding, trafficking, and degradation while minimizing misfolding and aggregation. Defective proteostasis leads to numerous protein misfolding diseases. Pharmacological chaperones are cell-permeant small molecules that promote the proper folding and trafficking of a protein via direct binding to that protein. They stabilize their target protein in a protein-pharmacological chaperone state, increasing the natively folded protein population that can effectively engage trafficking machinery for transport to the final destination for function. Here, as regards the application of pharmacological chaperones, we focus on their capability to promote the folding and trafficking of lysosomal enzymes, G protein coupled receptors (GPCRs), and ion channels, each of which is presently an important drug target. Pharmacological chaperones hold great promise as potential therapeutics to ameliorate a variety of protein misfolding diseases.


Subject(s)
Drug Discovery , Ion Channels/metabolism , Lysosomes/enzymology , Protein Folding/drug effects , Protein Transport/drug effects , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ion Channels/chemistry , Lysosomes/drug effects , Lysosomes/pathology , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Receptors, G-Protein-Coupled/chemistry
8.
Chem Biol ; 20(12): 1456-68, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24211135

ABSTRACT

GABA(A) receptors are the primary inhibitory ion channels in the mammalian central nervous system. The A322D mutation in the α1 subunit of GABA(A) receptors is known to result in its degradation and reduce its cell surface expression, leading to loss of GABAA receptor function in autosomal dominant juvenile myoclonic epilepsy. Here, we show that SAHA, a FDA-approved drug, increases the transcription of the α1(A322D) subunit, enhances its folding and trafficking posttranslationally, increases its cell surface level, and restores the GABA-induced maximal current in HEK293 cells expressing α1(A322D)ß2γ2 receptors to 10% of that for wild-type receptors. To enhance the trafficking efficiency of the α1(A322D) subunit, SAHA increases the BiP protein level and the interaction between the α1(A322D) subunit and calnexin. SAHA is a drug that enhances epilepsy-associated GABAA receptor proteostasis.


Subject(s)
Epilepsy/genetics , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Protein Transport/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Calnexin/metabolism , HEK293 Cells , Humans , Point Mutation , Protein Folding/drug effects , Protein Interaction Maps/drug effects , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/chemistry , Vorinostat , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...