Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2719, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38548794

RESUMO

The study of phosphorylase kinase (PhK)-regulated glycogen metabolism has contributed to the fundamental understanding of protein phosphorylation; however, the molecular mechanism of PhK remains poorly understood. Here we present the high-resolution cryo-electron microscopy structures of human muscle PhK. The 1.3-megadalton PhK α4ß4γ4δ4 hexadecamer consists of a tetramer of tetramer, wherein four αßγδ modules are connected by the central ß4 scaffold. The α- and ß-subunits possess glucoamylase-like domains, but exhibit no detectable enzyme activities. The α-subunit serves as a bridge between the ß-subunit and the γδ subcomplex, and facilitates the γ-subunit to adopt an autoinhibited state. Ca2+-free calmodulin (δ-subunit) binds to the γ-subunit in a compact conformation. Upon binding of Ca2+, a conformational change occurs, allowing for the de-inhibition of the γ-subunit through a spring-loaded mechanism. We also reveal an ADP-binding pocket in the ß-subunit, which plays a role in allosterically enhancing PhK activity. These results provide molecular insights of this important kinase complex.


Assuntos
Músculos , Fosforilase Quinase , Humanos , Fosforilase Quinase/química , Fosforilase Quinase/metabolismo , Microscopia Crioeletrônica , Subunidades Proteicas/metabolismo , Músculos/metabolismo
2.
Int J Biol Macromol ; 138: 181-187, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31279057

RESUMO

Many functions of phosphorylase kinase (PhK) are regulated by Ca2+ and Mg2+ ions. Ca2+ and Mg2+ ions stimulate activity of PhK, induce the changes in the tertiary and quaternary structure of the hexadecameric enzyme molecule, provoke association/aggregation of PhK molecules, enhance PhK binding to glycogen. To establish the kinetic regime of Ca2+ and Mg2+-induced aggregation of PhK from rabbit skeletal muscles at 40 °C, in the present work the kinetics of aggregation was studied at various protein concentrations using the dynamic light scattering. The proposed mechanism of aggregation involves the stage of unfolding of the protein molecule with retention of the integrity of its oligomeric structure, the nucleation stage and stages of the growth of protein aggregates. The initial rate of the aggregation process at the stage of aggregate growth depends linearly on the protein concentration. This means that the order of aggregation with respect to the protein is equal to unity and the aggregation rate is limited by the rate of protein unfolding. The rate constant of the first order characterizing the stage of protein unfolding was found to be equal to 0.071 min-1 (40 mM Hepes, pH 6.8, 100 mM NaCl, 0.1 mM Ca2+, 10 mM Mg2+).


Assuntos
Cálcio/farmacologia , Magnésio/farmacologia , Fosforilase Quinase/química , Agregados Proteicos/efeitos dos fármacos , Temperatura , Cinética , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína
3.
Genet Med ; 21(4): 772-789, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30659246

RESUMO

PURPOSE: Glycogen storage disease (GSD) types VI and IX are rare diseases of variable clinical severity affecting primarily the liver. GSD VI is caused by deficient activity of hepatic glycogen phosphorylase, an enzyme encoded by the PYGL gene. GSD IX is caused by deficient activity of phosphorylase kinase (PhK), the enzyme subunits of which are encoded by various genes: ɑ (PHKA1, PHKA2), ß (PHKB), É£ (PHKG1, PHKG2), and δ (CALM1, CALM2, CALM3). Glycogen storage disease types VI and IX have a wide spectrum of clinical manifestations and often cannot be distinguished from each other, or from other liver GSDs, on clinical presentation alone. Individuals with GSDs VI and IX can present with hepatomegaly with elevated serum transaminases, ketotic hypoglycemia, hyperlipidemia, and poor growth. This guideline for the management of GSDs VI and IX was developed as an educational resource for health-care providers to facilitate prompt and accurate diagnosis and appropriate management of patients. METHODS: A national group of experts in various aspects of GSDs VI and IX met to review the limited evidence base from the scientific literature and provided their expert opinions. Consensus was developed in each area of diagnosis, treatment, and management. Evidence bases for these rare disorders are largely based on expert opinion, particularly when targeted therapeutics that have to clear the US Food and Drug Administration (FDA) remain unavailable. RESULTS: This management guideline specifically addresses evaluation and diagnosis across multiple organ systems involved in GSDs VI and IX. Conditions to consider in a differential diagnosis stemming from presenting features and diagnostic algorithms are discussed. Aspects of diagnostic evaluation and nutritional and medical management, including care coordination, genetic counseling, and prenatal diagnosis are addressed. CONCLUSION: A guideline that will facilitate the accurate diagnosis and optimal management of patients with GSDs VI and IX was developed. This guideline will help health-care providers recognize patients with GSDs VI and IX, expedite diagnosis, and minimize adverse sequelae from delayed diagnosis and inappropriate management. It will also help identify gaps in scientific knowledge that exist today and suggest future studies.


Assuntos
Genômica , Doença de Depósito de Glicogênio/genética , Hipoglicemia/genética , Fosforilase Quinase/genética , Gerenciamento Clínico , Genética Médica/tendências , Glicogênio/genética , Glicogênio/metabolismo , Doença de Depósito de Glicogênio/diagnóstico , Doença de Depósito de Glicogênio/epidemiologia , Doença de Depósito de Glicogênio/terapia , Guias como Assunto , Humanos , Hipoglicemia/metabolismo , Hipoglicemia/terapia , Fígado/metabolismo , Fígado/patologia , Mutação , Fosforilase Quinase/química , Estados Unidos/epidemiologia
4.
J Biol Chem ; 293(19): 7099-7107, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29483194

RESUMO

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.


Assuntos
Encéfalo/enzimologia , Encéfalo/metabolismo , Glicogênio Fosforilase/metabolismo , Glicogenólise , Fosforilase Quinase/metabolismo , Animais , Metabolismo Energético , Glicogênio/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/química , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Glicogênio Fosforilase/química , Ensaios de Triagem em Larga Escala , Humanos , Isoenzimas/metabolismo , Ligantes , Fosforilase Quinase/química , Fosforilação , Relação Estrutura-Atividade , Transcriptoma
5.
Protein Sci ; 27(2): 472-484, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29098725

RESUMO

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.


Assuntos
Fosforilase Quinase/química , Subunidades Proteicas/química , Sítio Alostérico , Animais , Domínio Catalítico , Medição da Troca de Deutério , Glicogenólise , Humanos , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína
6.
Protein Sci ; 27(2): 485-497, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29098736

RESUMO

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.


Assuntos
Fosforilase Quinase/química , Subunidades Proteicas/química , Animais , Domínio Catalítico , Microscopia Crioeletrônica , Glicogenólise , Humanos , Modelos Moleculares , Multimerização Proteica , Estrutura Quaternária de Proteína
7.
Biochem Biophys Res Commun ; 482(2): 221-225, 2017 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-27845042

RESUMO

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.


Assuntos
Glicogênio Fosforilase/química , Fosforilase Quinase/química , Mapeamento de Interação de Proteínas/métodos , Sítios de Ligação , Reagentes de Ligações Cruzadas/química , Ativação Enzimática , Glicogênio Fosforilase/ultraestrutura , Complexos Multienzimáticos/química , Complexos Multienzimáticos/ultraestrutura , Fosforilase Quinase/ultraestrutura , Ligação Proteica , Subunidades Proteicas , Especificidade por Substrato
8.
Biochemistry ; 54(46): 6887-95, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26551836

RESUMO

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.


Assuntos
Fosforilase Quinase/química , Animais , Domínio Catalítico , Cristalografia por Raios X , Ativação Enzimática , Espectrometria de Massas , Modelos Moleculares , Mapeamento de Peptídeos , Fosforilase Quinase/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Subunidades Proteicas , Proteólise , Coelhos
9.
Protein Sci ; 24(12): 1956-63, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26362516

RESUMO

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.


Assuntos
Materiais Biomiméticos/química , Peptídeos/química , Fosforilase Quinase/química , Fosforilase Quinase/metabolismo , Sítios de Ligação , Domínio Catalítico , Ativação Enzimática , Glicogenólise , Modelos Moleculares , Complexos Multienzimáticos/química , Oxirredução , Fosforilação , Estrutura Secundária de Proteína
10.
J Mol Graph Model ; 61: 231-42, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26364215

RESUMO

Phosphorylase kinase (PhK) has been linked with a number of conditions such as glycogen storage diseases, psoriasis, type 2 diabetes and more recently, cancer (Camus et al., 2012 [6]). However, with few reported structural studies on PhK inhibitors, this hinders a structure based drug design approach. In this study, the inhibitory potential of 38 indirubin analogues have been investigated. 11 of these ligands had IC50 values in the range 0.170-0.360µM, with indirubin-3'-acetoxime (1c) the most potent. 7-Bromoindirubin-3'-oxime (13b), an antitumor compound which induces caspase-independent cell-death (Ribas et al., 2006 [20]) is revealed as a specific inhibitor of PhK (IC50=1.8µM). Binding assay experiments performed using both PhK-holo and PhK-γtrnc confirmed the inhibitory effects to arise from binding at the kinase domain (γ subunit). High level computations using QM/MM-PBSA binding free energy calculations were in good agreement with experimental binding data, as determined using statistical analysis, and support binding at the ATP-binding site. The value of a QM description for the binding of halogenated ligands exhibiting σ-hole effects is highlighted. A new statistical metric, the 'sum of the modified logarithm of ranks' (SMLR), has been defined which measures performance of a model for both the "early recognition" (ranking earlier/higher) of active compounds and their relative ordering by potency. Through a detailed structure activity relationship analysis considering other kinases (CDK2, CDK5 and GSK-3α/ß), 6'(Z) and 7(L) indirubin substitutions have been identified to achieve selective PhK inhibition. The key PhK binding site residues involved can also be targeted using other ligand scaffolds in future work.


Assuntos
Hipoglicemiantes/química , Indóis/química , Oximas/química , Fosforilase Quinase/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Motivos de Aminoácidos , Sítios de Ligação , Quinase 2 Dependente de Ciclina/química , Quinase 5 Dependente de Ciclina/química , Quinase 3 da Glicogênio Sintase/química , Glicogênio Sintase Quinase 3 beta , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Fosforilase Quinase/química , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Termodinâmica , Interface Usuário-Computador
11.
Int J Biol Macromol ; 68: 225-32, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24814653

RESUMO

Arginine is widely used in biotechnology as a folding enhancer and aggregation suppressor. However, its action on the stability of complexly organized oligomeric proteins, on the one hand, and its role in the formation of supramolecular structures, on the other hand, are poorly known. The investigation is concerned with the effects of arginine on protein-protein interactions using phosphorylase kinase (PhK) as an example. PhK, a 1.3MDa (αßγδ)4 hexadecameric complex, is a Ca(2+)-dependent regulatory enzyme that catalyzes phosphorylation and activation of glycogen phosphorylase b. On the basis of light scattering measurements it was shown that arginine induced aggregation of Ca(2+)-free PhK. On the contrary, when studying Ca(2+), Mg(2+)-induced aggregation of PhK at 37°C, the protective effect of arginine was demonstrated. The data on analytical ultracentrifugation are indicative of disruption of PhK hexadecameric structure under the action of arginine. Though HspB6 and HspB5 suppress aggregation of PhK they do not block the disruption effect of arginine with respect to both forms of PhK (Ca(2+)-free and Ca(2+), Mg(2+)-bound conformers). The dual effect of arginine has been interpreted from view-point of dual behaviour of arginine, functioning both like an osmolyte and a protein denaturant.


Assuntos
Arginina/farmacologia , Fosforilase Quinase/química , Agregados Proteicos/efeitos dos fármacos , Animais , Cálcio/metabolismo , Proteínas de Choque Térmico HSP20/metabolismo , Humanos , Hidrodinâmica , Íons , Cinética , Magnésio/farmacologia , Metilaminas/química , Fosforilase Quinase/metabolismo , Substâncias Protetoras/farmacologia , Coelhos , Temperatura , Ultracentrifugação
12.
Protein Sci ; 22(4): 444-54, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23359552

RESUMO

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.


Assuntos
Magnésio/química , Magnésio/metabolismo , Fosforilase Quinase/química , Fosforilase Quinase/metabolismo , Compostos Bicíclicos com Pontes/química , Compostos Bicíclicos com Pontes/metabolismo , Cátions/química , Cátions/metabolismo , Dicroísmo Circular , Dinitrofluorbenzeno/análogos & derivados , Dinitrofluorbenzeno/química , Dinitrofluorbenzeno/metabolismo , Luz , Conformação Proteica , Espalhamento de Radiação , Eletricidade Estática
13.
Mol Cell Proteomics ; 11(12): 1768-76, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964223

RESUMO

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.


Assuntos
Domínio Catalítico , Músculo Esquelético/enzimologia , Fosforilase Quinase , Subunidades Proteicas/análise , Catálise , Espectrometria de Massas , Músculo Esquelético/metabolismo , Fosforilase Quinase/análise , Fosforilase Quinase/química , Fosforilase Quinase/metabolismo , Fosforilação , Conformação Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas/química
14.
J Biol Chem ; 287(44): 36651-61, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22969083

RESUMO

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.


Assuntos
Fosforilase Quinase/química , Subunidades Proteicas/química , Sequência de Aminoácidos , Animais , Reagentes de Ligações Cruzadas/química , Dinitrofluorbenzeno/análogos & derivados , Dinitrofluorbenzeno/química , Simulação de Acoplamento Molecular , Fragmentos de Peptídeos/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Coelhos , Espectrometria de Massas em Tandem
15.
Adv Exp Med Biol ; 740: 703-30, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453966

RESUMO

Calcium/calmodulin-stimulated protein kinases can be classified as one of two types - restricted or multifunctional. This family of kinases contains several structural similarities: all possess a calmodulin binding motif and an autoinhibitory region. In addition, all of the calcium/calmodulin-stimulated protein kinases examined in this chapter are regulated by phosphorylation, which either activates or inhibits their kinase activity. However, as the multifunctional calcium/calmodulin-stimulated protein kinases are ubiquitously expressed, yet regulate a broad range of cellular functions, additional levels of regulation that control these cell-specific functions must exist. These additional layers of control include gene expression, signaling pathways, and expression of binding proteins and molecular targeting. All of the multifunctional calcium/calmodulin-stimulated protein kinases examined in this chapter appear to be regulated by these additional layers of control, however, this does not appear to be the case for the restricted kinases.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/química , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/química , Caseína Quinase I/síntese química , Caseína Quinase I/fisiologia , Humanos , Quinase de Cadeia Leve de Miosina/química , Quinase de Cadeia Leve de Miosina/fisiologia , Fosforilase Quinase/química , Fosforilase Quinase/fisiologia , Fosforilação
16.
Proteins ; 79(3): 703-19, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21287607

RESUMO

With an aim toward glycogenolysis control in Type 2 diabetes, we have investigated via kinetic experiments and computation the potential of indirubin (IC50 > 50 µM), indirubin-3'-oxime (IC50 = 144 nM), KT5720 (K(i) = 18.4 nM) and staurosporine (K(i) = 0.37 nM) as phosphorylase kinase (PhKγtrnc) ATP-binding site inhibitors, with the latter two revealed as potent inhibitors in the low nM range. Because of lack of structural information, we have exploited information from homologous kinase complexes to direct in silico calculations (docking, molecular dynamics, and MMGBSA) to predict the binding characteristics of the four ligands. All inhibitors are predicted to bind in the same active site area as the ATP adenine ring, with binding dominated by hinge region hydrogen bonds to Asp104:O and Met106:O (all four ligands) and also Met106:NH (for the indirubins). The PhKγtrnc-staurosporine complex has the greatest number of receptor-ligand hydrogen bonds, while for the indirubin-3'-oxime and KT5720 complexes there is an important network of interchanging water molecules bridging inhibitor-enzyme contacts. The MM-GBSA results revealed the source of staurosporine's low nM potency to be favorable electrostatic interactions, while KT5720 has strong van der Waals contributions. KT5720 interacts with the greatest number of protein residues either by direct or 1-water bridged hydrogen bond interactions, and the potential for more selective PhK inhibition based on a KT5720 analogue has been established. Including receptor flexibility in Schrödinger induced-fit docking calculations in most cases correctly predicted the binding modes as compared with the molecular dynamics structures; the algorithm was less effective when there were key structural waters bridging receptor-ligand contacts.


Assuntos
Trifosfato de Adenosina/antagonistas & inibidores , Carbazóis/química , Fosforilase Quinase/antagonistas & inibidores , Pirróis/química , Estaurosporina/química , Água/química , Trifosfato de Adenosina/química , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Indóis/química , Cinética , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Fosforilase Quinase/química , Homologia de Sequência de Aminoácidos
17.
Biochemistry ; 49(31): 6505-7, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20604537

RESUMO

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.


Assuntos
Acarbose/farmacologia , Glucana 1,4-alfa-Glucosidase/antagonistas & inibidores , Fosforilase Quinase/química , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos , Humanos , Hipoglicemiantes , Fosforilase Quinase/efeitos dos fármacos , Ligação Proteica , Conformação Proteica
18.
Biochemistry (Mosc) ; 74(5): 562-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19538131

RESUMO

Dynamic light scattering was used to study the interaction of phosphorylase kinase (PhK) and glycogen phosphorylase b (Phb) from rabbit skeletal muscle with glycogen under molecular crowding conditions arising from the presence of 1 M trimethylamine N-oxide and at physiological ionic strength. The mean value of hydrodynamic radius of the initial glycogen particles was 52 nm. Crowding stimulated Phb and PhK combined binding on glycogen particles. Two-stage character of PhK binding to glycogen particles containing adsorbed Phb was found in the presence of the crowding agent. At the initial stage, limited size particles with hydrodynamic radius of approximately 220 nm are formed, whereas the second stage is accompanied by linear growth of hydrodynamic radius. Flavin adenine dinucleotide (FAD) selectively inhibited PhK binding at the second stage. The data indicate that in the first stage Phb is involved in PhK binding by glycogen particles containing adsorbed Phb, whereas PhK binding in the second stage does not involve Phb.


Assuntos
Inibidores Enzimáticos/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Glicogênio Fosforilase Muscular/metabolismo , Glicogênio/metabolismo , Substâncias Macromoleculares/metabolismo , Fosforilase Quinase/metabolismo , Animais , Glicogênio Fosforilase Muscular/química , Substâncias Macromoleculares/química , Tamanho da Partícula , Fosforilase Quinase/química , Ligação Proteica , Coelhos
19.
Structure ; 17(1): 117-27, 2009 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-19141288

RESUMO

Phosphorylase kinase (PhK) coordinates hormonal and neuronal signals to initiate the breakdown of glycogen. The enzyme catalyzes the phosphorylation of inactive glycogen phosphorylase b (GPb), resulting in the formation of active glycogen phosphorylase a. We present a 9.9 angstroms resolution structure of PhK heterotetramer (alphabetagammadelta)4 determined by cryo-electron microscopy single-particle reconstruction. The enzyme has a butterfly-like shape comprising two lobes with 222 symmetry. This three-dimensional structure has allowed us to dock the catalytic gamma subunit to the PhK holoenzyme at a location that is toward the ends of the lobes. We have also determined the structure of PhK decorated with GPb at 18 angstroms resolution, which shows the location of the substrate near the kinase subunit. The PhK preparation contained a number of smaller particles whose structure at 9.8 angstroms resolution was consistent with a proteolysed activated form of PhK that had lost the alpha subunits and possibly the gamma subunits.


Assuntos
Glicogênio Fosforilase/metabolismo , Fosforilase Quinase/química , Domínio Catalítico , Cromatografia em Gel , Microscopia Crioeletrônica , Cristalografia por Raios X , Eletroforese em Gel de Poliacrilamida , Modelos Moleculares , Fosforilase Quinase/metabolismo , Conformação Proteica , Especificidade por Substrato
20.
Biochim Biophys Acta ; 1782(11): 664-70, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18950708

RESUMO

Mutations in the liver isoform of the Phosphorylase Kinase (PhK) alpha subunit (PHKA2 gene) cause X-linked liver glycogenosis (XLG), the most frequent type of PhK deficiency (glycogen-storage disease type IX). XLG patients can be divided in two subgroups, with similar clinical features but different activity of PhK (decreased in liver and blood cells for XLG-I and low in liver but normal or enhanced in blood cells for XLG-II). Here, we show that the PHKA2 missense mutations and small in-frame deletions/insertions are concentrated into two domains of the protein, which were recently described. In the N-terminal glucoamylase domain, mutations (principally leading to XLG-II) are clustered within the predicted glycoside-binding site, suggesting that they may have a direct impact on a possible hydrolytic activity of the PhK alpha subunit, which remains to be demonstrated. In the C-terminal calcineurin B-like domain (domain D), mutations (principally leading to XLG-I) are clustered in a region predicted to interact with the regulatory region of the PhK catalytic subunit and in a region covering this interaction site. Altogether, these results show that PHKA2 missense mutations or small in-frame deletions/insertions may have a direct impact on the PhK alpha functions and provide a framework for further experimental investigation.


Assuntos
Doença de Depósito de Glicogênio/enzimologia , Isoenzimas/genética , Mutação , Fosforilase Quinase/genética , Subunidades Proteicas/genética , Sequência de Aminoácidos , Análise Mutacional de DNA , Humanos , Isoenzimas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforilase Quinase/química , Fosforilase Quinase/metabolismo , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA