Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Jun 24.
Article in English | MEDLINE | ID: mdl-38913408

ABSTRACT

Allosteric cooperativity between ATP and substrates is a prominent characteristic of the cAMP-dependent catalytic subunit of protein kinase A (PKA-C). This long-range synergistic action is involved in substrate recognition and fidelity, and it may also regulate PKA's association with regulatory subunits and other binding partners. To date, a complete understanding of this intramolecular mechanism is still lacking. Here, we integrated NMR(Nuclear Magnetic Resonance)-restrained molecular dynamics simulations and a Markov State Model to characterize the free energy landscape and conformational transitions of PKA-C. We found that the apoenzyme populates a broad free energy basin featuring a conformational ensemble of the active state of PKA-C (ground state) and other basins with lower populations (excited states). The first excited state corresponds to a previously characterized inactive state of PKA-C with the αC helix swinging outward. The second excited state displays a disrupted hydrophobic packing around the regulatory (R) spine, with a flipped configuration of the F100 and F102 residues at the αC-ß4 loop. We validated the second excited state by analyzing the F100A mutant of PKA-C, assessing its structural response to ATP and substrate binding. While PKA-CF100A preserves its catalytic efficiency with Kemptide, this mutation rearranges the αC-ß4 loop conformation, interrupting the coupling of the two lobes and abolishing the allosteric binding cooperativity. The highly conserved αC-ß4 loop emerges as a pivotal element to control the synergistic binding of nucleotide and substrate, explaining how mutations or insertions near or within this motif affect the function and drug sensitivity in homologous kinases.


Subject(s)
Molecular Dynamics Simulation , Allosteric Regulation , Adenosine Triphosphate/metabolism , Catalytic Domain , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/genetics , Protein Conformation , Protein Binding , Nucleotides/metabolism , Substrate Specificity , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics
2.
bioRxiv ; 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37745542

ABSTRACT

Allosteric cooperativity between ATP and substrates is a prominent characteristic of the cAMP-dependent catalytic (C) subunit of protein kinase A (PKA). Not only this long-range synergistic action is involved in substrate recognition and fidelity, but it is likely to regulate PKA association with regulatory subunits and other binding partners. To date, a complete understanding of the molecular determinants for this intramolecular mechanism is still lacking. Here, we used an integrated NMR-restrained molecular dynamics simulations and a Markov Model to characterize the free energy landscape and conformational transitions of the catalytic subunit of protein kinase A (PKA-C). We found that the apo-enzyme populates a broad free energy basin featuring a conformational ensemble of the active state of PKA-C (ground state) and other basins with lower populations (excited states). The first excited state corresponds to a previously characterized inactive state of PKA-C with the αC helix swinging outward. The second excited state displays a disrupted hydrophobic packing around the regulatory (R) spine, with a flipped configuration of the F100 and F102 residues at the tip of the αC-ß4 loop. To experimentally validate the second excited state, we mutated F100 into alanine and used NMR spectroscopy to characterize the binding thermodynamics and structural response of ATP and a prototypical peptide substrate. While the activity of PKA-CF100A toward a prototypical peptide substrate is unaltered and the enzyme retains its affinity for ATP and substrate, this mutation rearranges the αC-ß4 loop conformation interrupting the allosteric coupling between nucleotide and substrate. The highly conserved αC-ß4 loop emerges as a pivotal element able to modulate the synergistic binding between nucleotide and substrate and may affect PKA signalosome. These results may explain how insertion mutations within this motif affect drug sensitivity in other homologous kinases.

3.
Biochim Biophys Acta Biomembr ; 1860(6): 1335-1341, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29501609

ABSTRACT

Approximately, 70% of the Ca2+ ion transport into the sarcoplasmic reticulum is catalyzed by the sarcoplasmic reticulum Ca2+-ATPase (SERCA), whose activity is endogenously regulated by phospholamban (PLN). PLN comprises a TM inhibitory region and a cytoplasmic regulatory region that harbors a consensus sequence for cAMP-dependent protein kinase (PKA). The inhibitory region binds the ATPase, reducing its apparent Ca2+ binding affinity. ß-adrenergic stimulation activates PKA, which phosphorylates PLN at Ser 16, reversing its inhibitory function. Mutations and post-translational modifications of PLN may lead to dilated cardiomyopathy (DCM) and heart failure. PLN's cytoplasmic region interconverts between a membrane-associated T state and a membrane-detached R state. The importance of these structural transitions on SERCA regulation is emerging, but the effects of natural occurring mutations and their relevance to the progression of heart disease are unclear. Here we use solid-state NMR spectroscopy to investigate the structural dynamics of two lethal PLN mutations, R9C and R25C, which lead to DCM. We found that the R25C mutant enhances the dynamics of PLN and shifts the conformational equilibrium toward the R state confirmation, whereas the R9C mutant drives the amphipathic cytoplasmic domain toward the membrane-associate state, enriching the T state population. The changes in membrane interactions caused by these mutations may explain the aberrant regulation of SERCA.


Subject(s)
Amino Acid Substitution , Calcium-Binding Proteins/chemistry , Cardiomyopathy, Dilated/genetics , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Mutation, Missense , Point Mutation , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Consensus Sequence , Humans , Membrane Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
4.
Biochim Biophys Acta ; 1848(1 Pt B): 315-22, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25251363

ABSTRACT

Phospholamban (PLN) is a single-pass membrane protein that regulates the sarco(endo)plasmic reticulum Ca²âº-ATPase (SERCA). Phosphorylation of PLN at Ser16 reverses its inhibitory function under ß-adrenergic stimulation, augmenting Ca²âº uptake in the sarcoplasmic reticulum and muscle contractility. PLN exists in two conformations; a T state, where the cytoplasmic domain is helical and adsorbed on the membrane surface, and an R state, where the cytoplasmic domain is unfolded and membrane detached. Previous studies have shown that the PLN conformational equilibrium is crucial to SERCA regulation. Here, we used a combination of solution and solid-state NMR to compare the structural topology and conformational dynamics of monomeric PLN (PLN(AFA)) with that of the PLN(R14del), a naturally occurring deletion mutant that is linked to the progression of dilated cardiomyopathy. We found that the behavior of the inhibitory transmembrane domain of PLN(R14del) is similar to that of the native sequence. Conversely, the conformational dynamics of R14del both in micelles and lipid membranes are enhanced. We conclude that the deletion of Arg14 in the cytoplasmic region weakens the interactions with the membrane and shifts the conformational equilibrium of PLN toward the disordered R state. This conformational transition is correlated with the loss-of-function character of this mutant and is corroborated by SERCA's activity assays. These findings support our hypothesis that SERCA function is fine-tuned by PLN conformational dynamics and begin to explain the aberrant regulation of SERCA by the R14del mutant.


Subject(s)
Arginine/chemistry , Calcium-Binding Proteins/chemistry , Membrane Proteins/chemistry , Lipid Bilayers/chemistry , Micelles , Protein Conformation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
5.
J Muscle Res Cell Motil ; 33(6): 485-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22971924

ABSTRACT

Phospholamban (PLN) is the endogenous inhibitor of the sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA), the integral membrane enzyme responsible for 70 % of the removal of Ca(2+) from the cytosol, inducing cardiac muscle relaxation in humans. Dysfunctions in SERCA:PLN interactions have been implicated as having a critical role in cardiac disease, and targeting Ca(2+) transport has been demonstrated to be a promising avenue in treating conditions of heart failure. Here, we designed a series of new mutants able to tune SERCA function, targeting the loop sequence that connects the transmembrane and cytoplasmic helices of PLN. We found that a variable degree of loss of inhibition mutants is attainable by engineering glycine mutations along PLN's loop domain. Remarkably, a double glycine mutation results in a complete loss-of-function mutant, fully mimicking the phosphorylated state of PLN. Using nuclear magnetic resonance spectroscopy, we rationalized the effects of these mutations in terms of entropic control on PLN function, whose inhibitory function can be modulated by increasing its conformational dynamics. However, if PLN mutations go past a threshold set by the phosphorylated state, they break the structural coupling between the transmembrane and cytoplasmic domains, resulting in a species that behaves as the inhibitory transmembrane domain alone. These studies provide new potential candidates for gene therapy to reverse the effects of heart failure.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cytoplasm/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Amino Acid Sequence , Glycine/genetics , Glycine/metabolism , Humans , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors
6.
Proc Natl Acad Sci U S A ; 108(7): 2735-40, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21282613

ABSTRACT

The regulatory interaction of phospholamban (PLN) with Ca(2+)-ATPase controls the uptake of calcium into the sarcoplasmic reticulum, modulating heart muscle contractility. A missense mutation in PLN cytoplasmic domain (R9C) triggers dilated cardiomyopathy in humans, leading to premature death. Using a combination of biochemical and biophysical techniques both in vitro and in live cells, we show that the R9C mutation increases the stability of the PLN pentameric assembly via disulfide bridge formation, preventing its binding to Ca(2+)-ATPase as well as phosphorylation by protein kinase A. These effects are enhanced under oxidizing conditions, suggesting that oxidative stress may exacerbate the cardiotoxic effects of the PLN(R9C) mutant. These results reveal a regulatory role of the PLN pentamer in calcium homeostasis, going beyond the previously hypothesized role of passive storage for active monomers.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Mutation, Missense/genetics , Myocardial Contraction/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Cardiomyopathy, Dilated/metabolism , Fluorescence Resonance Energy Transfer , Humans , Myocardial Contraction/physiology , Oxidative Stress/genetics , Phosphorylation
7.
Methods Mol Biol ; 654: 303-19, 2010.
Article in English | MEDLINE | ID: mdl-20665273

ABSTRACT

This chapter reviews the molecular biology, biochemical, and NMR methods that we used to study the structural dynamics, membrane topology, and interaction of phospholamban (PLN), a small regulatory membrane protein involved in the regulation of the sarcoplasmic reticulum Ca-ATPase (SERCA). In particular, we show the progression of our research from the initial hypotheses toward understanding the molecular mechanisms of SERCA's regulation, including the effects of PLN oligomerization and posttranslational phosphorylation. Finally, we show how the knowledge of the molecular mechanism of the structural dynamics and topology of free and bound proteins can lead to the rational design of PLN analogs for possible use in gene therapy.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Animals , Calcium-Binding Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Multimerization , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
Protein Expr Purif ; 64(2): 231-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19027069

ABSTRACT

Extensive X-ray crystallographic studies carried out on the catalytic-subunit of protein kinase A (PKA-C) enabled the atomic characterization of inhibitor and/or substrate peptide analogues trapped at its active site. Yet, the structural and dynamic transitions of these peptides from the free to the bound state are missing. These conformational transitions are central to understanding molecular recognition and the enzymatic cycle. NMR spectroscopy allows one to study these phenomena under functionally relevant conditions. However, the amounts of isotopically labeled peptides required for this technique present prohibitive costs for solid-phase peptide synthesis. To enable NMR studies, we have optimized both expression and purification of isotopically enriched substrate/inhibitor peptides using a recombinant fusion protein system. Three of these peptides correspond to the cytoplasmic regions of the wild-type and lethal mutants of the membrane protein phospholamban, while the fourth peptide correspond to the binding epitope of the heat-stable protein kinase inhibitor (PKI(5-24)). The target peptides were fused to the maltose binding protein (MBP), which is further purified using a His(6) tag approach. This convenient protocol allows for the purification of milligram amounts of peptides necessary for NMR analysis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Peptide Fragments/chemistry , Amino Acid Sequence , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Catalytic Domain , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Isotope Labeling , Magnetic Resonance Spectroscopy , Maltose-Binding Proteins , Models, Genetic , Molecular Sequence Data , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity
9.
Biochemistry ; 47(1): 3-13, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18081313

ABSTRACT

Phospholamban (PLN) and sarcolipin (SLN) are two single-pass membrane proteins that regulate Ca2+-ATPase (SERCA), an ATP-driven pump that translocates calcium ions into the lumen of the sarcoplasmic reticulum, initiating muscle relaxation. Both proteins bind SERCA through intramembrane interactions, impeding calcium translocation. While phosphorylation of PLN at Ser-16 and/or Thr-17 reestablishes calcium flux, the regulatory mechanism of SLN remains elusive. SERCA has been crystallized in several different states along the enzymatic reaction coordinates, providing remarkable mechanistic information; however, the lack of high-resolution crystals in the presence of PLN and SLN limits the current understanding of the regulatory mechanism. This brief review offers a survey of our hybrid structural approach using solution and solid-state NMR methodologies to understand SERCA regulation from the point of view of PLN and SLN. These results have improved our understanding of the calcium translocation process and are the basis for designing new therapeutic approaches to ameliorate muscle malfunctions.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Transporting ATPases/chemistry , Muscle Proteins/chemistry , Proteolipids/chemistry , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Muscle Proteins/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proteolipids/metabolism
10.
J Biol Chem ; 282(51): 37205-14, 2007 Dec 21.
Article in English | MEDLINE | ID: mdl-17908690

ABSTRACT

Cardiac contraction and relaxation are regulated by conformational transitions of protein complexes that are responsible for calcium trafficking through cell membranes. Central to the muscle relaxation phase is a dynamic membrane protein complex formed by Ca2+-ATPase (SERCA) and phospholamban (PLN), which in humans is responsible for approximately 70% of the calcium re-uptake in the sarcoplasmic reticulum. Dysfunction in this regulatory mechanism causes severe pathophysiologies. In this report, we used a combination of nuclear magnetic resonance, electron paramagnetic resonance, and coupled enzyme assays to investigate how single mutations at position 21 of PLN affects its structural dynamics and, in turn, its interaction with SERCA. We found that it is possible to control the activity of SERCA by tuning PLN structural dynamics. Both increased rigidity and mobility of the PLN backbone cause a reduction of SERCA inhibition, affecting calcium transport. Although the more rigid, loss-of-function (LOF) mutants have lower binding affinities for SERCA, the more dynamic LOF mutants have binding affinities similar to that of PLN. Here, we demonstrate that it is possible to harness this knowledge to design new LOF mutants with activity similar to S16E (a mutant already used in gene therapy) for possible application in recombinant gene therapy. As proof of concept, we show a new mutant of PLN, P21G, with improved LOF characteristics in vitro.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cell Membrane/enzymology , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Amino Acid Substitution , Animals , Calcium/chemistry , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Electron Spin Resonance Spectroscopy , Genetic Therapy , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/physiopathology , Heart Diseases/therapy , Humans , Ion Transport/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation, Missense , Myocardial Contraction/genetics , Nuclear Magnetic Resonance, Biomolecular , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...