Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Adv Neurobiol ; 23: 1-16, 2019.
Article in English | MEDLINE | ID: mdl-31667804

ABSTRACT

Brain glycogen is extremely difficult to study because it is very labile to physiological status and postmortem autolysis, and glycogen degradative enzymes are rapidly activated by metabolites and signaling molecules. Glycogen is predominantly located within astrocytes in adult brain, and abnormal glycogen metabolism in neurons has lethal consequences. Diverse distribution of glycogen among subcellular compartments suggests local regulation and different functional roles, and recent studies have revealed critically important roles for glycogen in normal brain function and Lafora disease. This brief overview highlights some of the major advances in elucidation of glycogen's roles in astrocytic functions and neurotransmission and the severe consequences of aberrant neuronal glycogen metabolism.


Subject(s)
Brain/physiology , Energy Metabolism , Glycogen/metabolism , Astrocytes/metabolism , Biomedical Research , Brain/cytology , Brain/metabolism , Humans , Lafora Disease/metabolism , Neurons/metabolism
2.
J Biol Chem ; 293(19): 7087-7088, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29514979

ABSTRACT

The synthesis of glycogen allows for efficient intracellular storage of glucose molecules in a soluble form that can be rapidly released to enter glycolysis in response to energy demand. Intensive studies of glucose and glycogen metabolism, predominantly in skeletal muscle and liver, have produced innumerable insights into the mechanisms of hormone action, resulting in the award of several Nobel Prizes over the last one hundred years. Glycogen is actually present in all cells and tissues, albeit at much lower levels than found in muscle or liver. However, metabolic and physiological roles of glycogen in other tissues are poorly understood. This series of Minireviews summarizes what is known about the enzymes involved in brain glycogen metabolism and studies that have linked glycogen metabolism to multiple brain functions involving metabolic communication between astrocytes and neurons. Recent studies unexpectedly linking some forms of epilepsy to mutations in two poorly understood proteins involved in glycogen metabolism are also reviewed.


Subject(s)
Brain/metabolism , Glycogen/metabolism , Brain/enzymology , Glycogen/biosynthesis , Glycogenolysis , Glycolysis , Humans , Review Literature as Topic , Synaptic Transmission
3.
J Biol Chem ; 293(19): 7099-7107, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29483194

ABSTRACT

The key regulatory enzymes of glycogenolysis are phosphorylase kinase, a hetero-oligomer with four different types of subunits, and glycogen phosphorylase, a homodimer. Both enzymes are activated by phosphorylation and small ligands, and both enzymes have distinct isoforms that are predominantly expressed in muscle, liver, or brain; however, whole-transcriptome high-throughput sequencing analyses show that in brain both of these enzymes are likely composed of subunit isoforms representing all three tissues. This Minireview examines the regulatory properties of the isoforms of these two enzymes expressed in the three tissues, focusing on their potential regulatory similarities and differences. Additionally, the activity, structure, and regulation of the remaining enzyme necessary for glycogenolysis, glycogen-debranching enzyme, are also reviewed.


Subject(s)
Brain/enzymology , Brain/metabolism , Glycogen Phosphorylase/metabolism , Glycogenolysis , Phosphorylase Kinase/metabolism , Animals , Energy Metabolism , Glycogen/metabolism , Glycogen Debranching Enzyme System/chemistry , Glycogen Debranching Enzyme System/metabolism , Glycogen Phosphorylase/chemistry , High-Throughput Screening Assays , Humans , Isoenzymes/metabolism , Ligands , Phosphorylase Kinase/chemistry , Phosphorylation , Structure-Activity Relationship , Transcriptome
4.
Protein Sci ; 27(2): 472-484, 2018 02.
Article in English | MEDLINE | ID: mdl-29098725

ABSTRACT

Phosphorylase kinase (PhK), a 1.3 MDa regulatory enzyme complex in the glycogenolysis cascade, has four copies each of four subunits, (αßγδ)4 , and 325 kDa of unique sequence (the mass of an αßγδ protomer). The α, ß and δ subunits are regulatory, and contain allosteric activation sites that stimulate the activity of the catalytic γ subunit in response to diverse signaling molecules. Due to its size and complexity, no high resolution structures have been solved for the intact complex or its regulatory α and ß subunits. Of PhK's four subunits, the least is known about the structure and function of its largest subunit, α. Here, we have modeled the full-length α subunit, compared that structure against previously predicted domains within this subunit, and performed hydrogen-deuterium exchange on the intact subunit within the PhK complex. Our modeling results show α to comprise two major domains: an N-terminal glycoside hydrolase domain and a large C-terminal importin α/ß-like domain. This structure is similar to our previously published model for the homologous ß subunit, although clear structural differences are present. The overall highly helical structure with several intervening hinge regions is consistent with our hydrogen-deuterium exchange results obtained for this subunit as part of the (αßγδ)4 PhK complex. Several low exchanging regions predicted to lack ordered secondary structure are consistent with inter-subunit contact sites for α in the quaternary structure of PhK; of particular interest is a low-exchanging region in the C-terminus of α that is known to bind the regulatory domain of the catalytic γ subunit.


Subject(s)
Phosphorylase Kinase/chemistry , Protein Subunits/chemistry , Allosteric Site , Animals , Catalytic Domain , Deuterium Exchange Measurement , Glycogenolysis , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Quaternary , Protein Structure, Secondary
5.
Protein Sci ; 27(2): 485-497, 2018 02.
Article in English | MEDLINE | ID: mdl-29098736

ABSTRACT

In the tightly regulated glycogenolysis cascade, the breakdown of glycogen to glucose-1-phosphate, phosphorylase kinase (PhK) plays a key role in regulating the activity of glycogen phosphorylase. PhK is a 1.3 MDa hexadecamer, with four copies each of four different subunits (α, ß, γ and δ), making the study of its structure challenging. Using hydrogen-deuterium exchange, we have analyzed the regulatory ß subunit and the catalytic γ subunit in the context of the intact non-activated PhK complex to study the structure of these subunits and identify regions of surface exposure. Our data suggest that within the non-activated complex the γ subunit assumes an activated conformation and are consistent with a previous docking model of the ß subunit within the cryoelectron microscopy envelope of PhK.


Subject(s)
Phosphorylase Kinase/chemistry , Protein Subunits/chemistry , Animals , Catalytic Domain , Cryoelectron Microscopy , Glycogenolysis , Humans , Models, Molecular , Protein Multimerization , Protein Structure, Quaternary
6.
Biochem Biophys Res Commun ; 482(2): 221-225, 2017 Jan 08.
Article in English | MEDLINE | ID: mdl-27845042

ABSTRACT

The selective phosphorylation of glycogen phosphorylase (GP) by its only known kinase, phosphorylase kinase (PhK), keeps glycogen catabolism tightly regulated. In addition to the obligatory interaction between the catalytic γ subunit of PhK and the phosphorylatable region of GP, previous studies have suggested additional sites of interaction between this kinase and its protein substrate. Using short chemical crosslinkers, we have identified direct interactions of GP with the large regulatory α and ß subunits of PhK. These newfound interactions were found to be sensitive to ligands that bind PhK.


Subject(s)
Glycogen Phosphorylase/chemistry , Phosphorylase Kinase/chemistry , Protein Interaction Mapping/methods , Binding Sites , Cross-Linking Reagents/chemistry , Enzyme Activation , Glycogen Phosphorylase/ultrastructure , Multienzyme Complexes/chemistry , Multienzyme Complexes/ultrastructure , Phosphorylase Kinase/ultrastructure , Protein Binding , Protein Subunits , Substrate Specificity
7.
Adv Exp Med Biol ; 919: 397-431, 2016.
Article in English | MEDLINE | ID: mdl-27975228

ABSTRACT

Modern mass spectrometry (MS) technologies have provided a versatile platform that can be combined with a large number of techniques to analyze protein structure and dynamics. These techniques include the three detailed in this chapter: (1) hydrogen/deuterium exchange (HDX), (2) limited proteolysis, and (3) chemical crosslinking (CX). HDX relies on the change in mass of a protein upon its dilution into deuterated buffer, which results in varied deuterium content within its backbone amides. Structural information on surface exposed, flexible or disordered linker regions of proteins can be achieved through limited proteolysis, using a variety of proteases and only small extents of digestion. CX refers to the covalent coupling of distinct chemical species and has been used to analyze the structure, function and interactions of proteins by identifying crosslinking sites that are formed by small multi-functional reagents, termed crosslinkers. Each of these MS applications is capable of revealing structural information for proteins when used either with or without other typical high resolution techniques, including NMR and X-ray crystallography.


Subject(s)
Computational Biology/methods , Data Mining/methods , Databases, Protein , Mass Spectrometry/methods , Proteins/analysis , Proteome , Proteomics/methods , Algorithms , Animals , Cross-Linking Reagents/chemistry , Deuterium Exchange Measurement , High-Throughput Screening Assays , Humans , Protein Conformation , Proteolysis , Reproducibility of Results , Software , Workflow
8.
Biophys J ; 110(12): 2809, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332139
9.
Biophys J ; 110(9): 1912-23, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27166800

ABSTRACT

Allosteric regulation of protein function is recognized to be widespread throughout biology; however, knowledge of allosteric mechanisms, the molecular changes within a protein that couple one binding site to another, is limited. Although mutagenesis is often used to probe allosteric mechanisms, we consider herein what the outcome of a mutagenesis study truly reveals about an allosteric mechanism. Arguably, the best way to evaluate the effects of a mutation on allostery is to monitor the allosteric coupling constant (Qax), a ratio of the substrate binding constants in the absence versus presence of an allosteric effector. A range of substitutions at a given residue position in a protein can reveal when a particular substitution causes gain-of-function, which addresses a key challenge in interpreting mutation-dependent changes in the magnitude of Qax. Thus, whole-protein mutagenesis studies offer an acceptable means of identifying residues that contribute to an allosteric mechanism. With this focus on monitoring Qax, and keeping in mind the equilibrium nature of allostery, we consider alternative possibilities for what an allosteric mechanism might be. We conclude that different possible mechanisms (rotation-of-solid-domains, movement of secondary structure, side-chain repacking, changes in dynamics, etc.) will result in different findings in whole-protein mutagenesis studies.


Subject(s)
Mutagenesis , Proteins/chemistry , Proteins/genetics , Allosteric Regulation , Models, Molecular , Mutation , Protein Conformation , Proteins/metabolism , Thermodynamics
10.
Biochemistry ; 54(51): 7524-30, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26632861

ABSTRACT

In the six decades since its discovery, phosphorylase kinase (PhK) from rabbit skeletal muscle has usually been studied at 30 °C; in fact, not a single study has examined functions of PhK at a rabbit's body temperature, which is nearly 10 °C greater. Thus, we have examined aspects of the activity, regulation, and structure of PhK at temperatures between 0 and 40 °C. Between 0 and 30 °C, the activity at pH 6.8 of nonphosphorylated PhK predictably increased; however, between 30 and 40 °C, there was a dramatic jump in its activity, resulting in the nonactivated enzyme having a far greater activity at body temperature than was previously realized. This anomalous change in properties between 30 and 40 °C was observed for multiple functions, and both stimulation (by ADP and phosphorylation) and inhibition (by orthophosphate) were considerably less pronounced at 40 °C than at 30 °C. In general, the allosteric control of PhK's activity is definitely more subtle at body temperature. Changes in behavior related to activity at 40 °C and its control can be explained by the near disappearance of hysteresis at physiological temperature. In important ways, the picture of PhK that has emerged from six decades of study at temperatures of ≤30 °C does not coincide with that of the enzyme studied at physiological temperature. The probable underlying mechanism for the dramatic increase in PhK's activity between 30 and 40 °C is an abrupt change in the conformations of the regulatory ß and catalytic γ subunits between these two temperatures.


Subject(s)
Body Temperature , Phosphorylase Kinase/metabolism , Animals , Enzyme Activation , Female , Phosphorylation , Rabbits
11.
Biochemistry ; 54(46): 6887-95, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26551836

ABSTRACT

Phosphorylase kinase (PhK) is a 1.3 MDa (αßγδ)4 enzyme complex, in which αßγδ protomers associate in D2 symmetry to form two large octameric lobes that are interconnected by four bridges. The approximate locations of the subunits have been mapped in low-resolution cryo-electron microscopy structures of the complex; however, the disposition of the subunits within the complex remains largely unknown. We have used partial proteolysis and chemical footprinting in combination with high-resolution mass spectrometry to identify surface-exposed regions of the intact nonactivated and phospho-activated conformers. In addition to the known interaction of the γ subunit's C-terminal regulatory domain with the δ subunit (calmodulin), our exposure results indicate that the catalytic core of γ may also anchor to the PhK complex at the bottom backside of its C-terminal lobe facing away from the active site cleft. Exposed loops on the α and ß regulatory subunits within the complex occur at regions overlapping with tissue-specific alternative RNA splice sites and regulatory phosphorylatable domains. Their phosphorylation alters the surface exposure of α and ß, corroborating previous biophysical and biochemical studies that detected phosphorylation-dependent conformational changes in these subunits; however, for the first time, specific affected regions have been identified.


Subject(s)
Phosphorylase Kinase/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Mass Spectrometry , Models, Molecular , Peptide Mapping , Phosphorylase Kinase/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Subunits , Proteolysis , Rabbits
12.
Protein Sci ; 24(12): 1956-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26362516

ABSTRACT

Phosphorylase kinase (PhK) is a hexadecameric (αßγδ)(4) enzyme complex that upon activation by phosphorylation stimulates glycogenolysis. Due to its large size (1.3 MDa), elucidating the structural changes associated with the activation of PhK has been challenging, although phosphoactivation has been linked with an increased tendency of the enzyme's regulatory ß-subunits to self-associate. Here we report the effect of a peptide mimetic of the phosphoryltable N-termini of ß on the selective, zero-length, oxidative crosslinking of these regulatory subunits to form ß-ß dimers in the nonactivated PhK complex. This peptide stimulated ß-ß dimer formation when not phosphorylated, but was considerably less effective in its phosphorylated form. Because this peptide mimetic of ß competes with its counterpart region in the nonactivated enzyme complex in binding to the catalytic γ-subunit, we were able to formulate a structural model for the phosphoactivation of PhK. In this model, the nonactivated state of PhK is maintained by the interaction between the nonphosphorylated N-termini of ß and the regulatory C-terminal domains of the γ-subunits; phosphorylation of ß weakens this interaction, leading to activation of the γ-subunits.


Subject(s)
Biomimetic Materials/chemistry , Peptides/chemistry , Phosphorylase Kinase/chemistry , Phosphorylase Kinase/metabolism , Binding Sites , Catalytic Domain , Enzyme Activation , Glycogenolysis , Models, Molecular , Multienzyme Complexes/chemistry , Oxidation-Reduction , Phosphorylation , Protein Structure, Secondary
13.
Protein Sci ; 22(4): 444-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23359552

ABSTRACT

For over four decades free Mg(2+) ions, that is, those in excess of MgATP, have been reported to affect a wide variety of properties of phosphorylase kinase (PhK), including its affinity for other molecules, proteolysis, chemical crosslinking, phosphorylation, binding to certain monoclonal antibodies, and activity, which is stimulated. Additionally, for over three decades Mg(2+) has been known to act synergistically with Ca(2+) , another divalent activator of PhK, to affect even more properties of the enzyme. During all of this time, however, no study has been performed to determine the overall effects of free Mg(2+) ions on the physical properties of PhK, even though the effects of Ca(2+) ions on PhK's properties are well documented. In this study, changes in the physicochemical properties of PhK induced by Mg(2+) under nonactivating (pH 6.8) and activating (pH 8.2) conditions were investigated by circular dichroism spectroscopy, zeta potential analyses, dynamic light scattering, second derivative UV absorption, negative stain electron microscopy, and differential chemical crosslinking. The effects of the activator Mg(2+) on some of the properties of PhK measured by these techniques were found to be quite different at the two pH values, and displayed both differences and similarities with the effects previously reported to be induced by the activator Ca(2+) (Liu et al., Protein Sci 2008;17:2111-2119). The similarities may reflect the fact that both cations are activators, and foremost among their similarities is the dramatically less negative zeta potential induced by their binding to PhK.


Subject(s)
Magnesium/chemistry , Magnesium/metabolism , Phosphorylase Kinase/chemistry , Phosphorylase Kinase/metabolism , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/metabolism , Cations/chemistry , Cations/metabolism , Circular Dichroism , Dinitrofluorobenzene/analogs & derivatives , Dinitrofluorobenzene/chemistry , Dinitrofluorobenzene/metabolism , Light , Protein Conformation , Scattering, Radiation , Static Electricity
14.
Mol Cell Proteomics ; 11(12): 1768-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22964223

ABSTRACT

Phosphorylase kinase (PhK), a 1.3 MDa enzyme complex that regulates glycogenolysis, is composed of four copies each of four distinct subunits (α, ß, γ, and δ). The catalytic protein kinase subunit within this complex is γ, and its activity is regulated by the three remaining subunits, which are targeted by allosteric activators from neuronal, metabolic, and hormonal signaling pathways. The regulation of activity of the PhK complex from skeletal muscle has been studied extensively; however, considerably less is known about the interactions among its subunits, particularly within the non-activated versus activated forms of the complex. Here, nanoelectrospray mass spectrometry and partial denaturation were used to disrupt PhK, and subunit dissociation patterns of non-activated and phospho-activated (autophosphorylation) conformers were compared. In so doing, we have established a network of subunit contacts that complements and extends prior evidence of subunit interactions obtained from chemical crosslinking, and these subunit interactions have been modeled for both conformers within the context of a known three-dimensional structure of PhK solved by cryoelectron microscopy. Our analyses show that the network of contacts among subunits differs significantly between the nonactivated and phospho-activated conformers of PhK, with the latter revealing new interprotomeric contact patterns for the ß subunit, the predominant subunit responsible for PhK's activation by phosphorylation. Partial disruption of the phosphorylated conformer yields several novel subcomplexes containing multiple ß subunits, arguing for their self-association within the activated complex. Evidence for the theoretical αßγδ protomeric subcomplex, which has been sought but not previously observed, was also derived from the phospho-activated complex. In addition to changes in subunit interaction patterns upon phospho-activation, mass spectrometry revealed a large change in the overall stability of the complex, with the phospho-activated conformer being more labile, in concordance with previous hypotheses on the mechanism of allosteric activation of PhK through perturbation of its inhibitory quaternary structure.


Subject(s)
Catalytic Domain , Muscle, Skeletal/enzymology , Phosphorylase Kinase , Protein Subunits/analysis , Catalysis , Mass Spectrometry , Muscle, Skeletal/metabolism , Phosphorylase Kinase/analysis , Phosphorylase Kinase/chemistry , Phosphorylase Kinase/metabolism , Phosphorylation , Protein Conformation , Protein Structure, Quaternary , Protein Subunits/chemistry
15.
J Biol Chem ; 287(44): 36651-61, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22969083

ABSTRACT

Phosphorylase kinase (PhK) is a hexadecameric (αßγδ)(4) complex that regulates glycogenolysis in skeletal muscle. Activity of the catalytic γ subunit is regulated by allosteric activators targeting the regulatory α, ß, and δ subunits. Three-dimensional EM reconstructions of PhK show it to be two large (αßγδ)(2) lobes joined with D(2) symmetry through interconnecting bridges. The subunit composition of these bridges was unknown, although indirect evidence suggested the ß subunits may be involved in their formation. We have used biochemical, biophysical, and computational approaches to not only address the quaternary structure of the ß subunits within the PhK complex, i.e. whether they compose the bridges, but also their secondary and tertiary structures. The secondary structure of ß was determined to be predominantly helical by comparing the CD spectrum of an αγδ subcomplex with that of the native (αßγδ)(4) complex. An atomic model displaying tertiary structure for the entire ß subunit was constructed using chemical cross-linking, MS, threading, and ab initio approaches. Nearly all this model is covered by two templates corresponding to glycosyl hydrolase 15 family members and the A subunit of protein phosphatase 2A. Regarding the quaternary structure of the ß subunits, they were directly determined to compose the four interconnecting bridges in the (αßγδ)(4) kinase core, because a ß(4) subcomplex was observed through both chemical cross-linking and top-down MS of PhK. The predicted model of the ß subunit was docked within the bridges of a cryoelectron microscopic density envelope of PhK utilizing known surface features of the subunit.


Subject(s)
Phosphorylase Kinase/chemistry , Protein Subunits/chemistry , Amino Acid Sequence , Animals , Cross-Linking Reagents/chemistry , Dinitrofluorobenzene/analogs & derivatives , Dinitrofluorobenzene/chemistry , Molecular Docking Simulation , Peptide Fragments/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary , Rabbits , Tandem Mass Spectrometry
16.
Methods Mol Biol ; 796: 117-32, 2012.
Article in English | MEDLINE | ID: mdl-22052488

ABSTRACT

This chapter explores the structural responses of a massive, hetero-oligomeric protein complex to a single allosteric activator as probed by a wide range of chemical, biochemical, and biophysical approaches. Some of the approaches used are amenable only to large protein targets, whereas others push the limits of their utility. Some of the techniques focus on individual subunits, or portions thereof, while others examine the complex as a whole. Despite the absence of crystallographic data for the complex, the diverse techniques identify and implicate a small region of its catalytic subunit as the master allosteric activation switch for the entire complex.


Subject(s)
Proteins/chemistry , Allosteric Regulation/physiology , Calcium/pharmacology , Fluorescence Resonance Energy Transfer , Immunochemistry , Microscopy, Electron , Protein Conformation/drug effects , Protein Multimerization , Proteins/metabolism , Proteins/ultrastructure
17.
Biochemistry ; 49(31): 6505-7, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20604537

ABSTRACT

Phosphorylase kinase (PhK), an (alphabetagammadelta)(4) complex, stimulates energy production from glycogen in the cascade activation of glycogenolysis. Its large homologous alpha and beta subunits regulate the activity of the catalytic gamma subunit and account for 81% of PhK's mass. Both subunits are thought to be multidomain structures, and recent predictions based on their sequences suggest the presence of potentially functional glucoamylase (GH15)-like domains near their amino termini. We present the first experimental evidence of such a domain in PhK by demonstrating that the glucoamylase inhibitor acarbose binds PhK, perturbs its structure, and stimulates its kinase activity.


Subject(s)
Acarbose/pharmacology , Glucan 1,4-alpha-Glucosidase/antagonists & inhibitors , Phosphorylase Kinase/chemistry , Enzyme Activation/drug effects , Enzyme Inhibitors , Humans , Hypoglycemic Agents , Phosphorylase Kinase/drug effects , Protein Binding , Protein Conformation
18.
IUBMB Life ; 62(2): 158-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19937969
19.
Biochemistry ; 48(42): 10183-91, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19764815

ABSTRACT

Understanding the regulatory interactions among the 16 subunits of the (alphabetagammadelta)(4) phosphorylase b kinase (PhK) complex can only be achieved through reconstructing the holoenzyme or its subcomplexes from the individual subunits. In this study, recombinant baculovirus carrying a vector containing a multigene cassette was created to coexpress in insect cells alpha, beta, gamma, and delta subunits corresponding to rabbit skeletal muscle PhK. The hexadecameric recombinant PhK (rPhK) and its corresponding alphagammadelta trimeric subcomplex were purified to homogeneity with proper subunit stoichiometries. The catalytic activity of rPhK at pH 8.2 and its ratio of activities at pH 6.8 versus pH 8.2 were comparable to those of PhK purified from rabbit muscle (RM PhK), as was the hysteresis (autoactivation) in the rate of product formation at pH 6.8. Both the rPhK and alphagammadelta exhibited only a very low Ca(2+)-independent activity and a Ca(2+)-dependent activity similar to that of the native holoenzyme with [Ca(2+)](0.5) of 0.4 microM for the RM PhK, 0.7 microM for the rPhK, and 1.5 microM for the alphagammadelta trimer. The RM PhK, rPhK, and alphagammadelta subcomplex were also all activated through self-phosphorylation. Using cross-linking and limited proteolysis, the alpha-gamma intersubunit contacts previously observed within the intact RM PhK complex were also observed within the recombinant alphagammadelta subcomplex. Our results indicate that both the rPhK and alphagammadelta subcomplex are promising models for future structure-function studies on the regulation of PhK activity through intersubunit contacts, because both retained the regulatory properties of the enzyme purified from skeletal muscle.


Subject(s)
Muscle, Skeletal/enzymology , Phosphorylase Kinase/metabolism , Protein Subunits/metabolism , Animals , Baculoviridae/metabolism , Holoenzymes/chemistry , Holoenzymes/metabolism , Kinetics , Models, Animal , Muscle, Skeletal/metabolism , Phosphorylation , Protein Subunits/chemistry , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...