Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 584
Filtrar
1.
J Nat Prod ; 86(11): 2562-2570, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-37906816

RESUMO

Four new natural chemical entities, including 2-hydroxy-α-truxillic acid (2), (3R,4S)-2,2-dimethyl-3-hydroxy-4-(1-angeloyloxy)-6-acetyl-7-methoxychromane (3), N-tricosanoyltyramine (4), and grandifolamide (5), were isolated along with 11 known compounds (1, 6-15) from the aerial parts of Ageratina grandifolia. The chemical structures were elucidated using chemical derivatization and HR-MS, NMR, and DFT-calculated chemical shifts, combined with DP4+ statistical analysis. It was found that 2 decomposed into its biogenetic precursor, o-coumaric acid, upon standing at room temperature for a few weeks. 3,5-Diprenyl-4-hydroxyacetophenone (8), O-methylencecalinol (10), encecalin (11), and encecalinol (12) bound to calmodulin (CaM) with higher affinity than chlorpromazine, a well-known CaM inhibitor. Molecular dynamics studies revealed that the complexes of these compounds with CaM remained stable during the simulation. Altogether these results revealed the therapeutic and research tool potential of compounds 8, 10, 11, and 12.


Assuntos
Ageratina , Ageratina/química , Calmodulina/química , Calmodulina/metabolismo , Calmodulina/farmacologia , Simulação de Dinâmica Molecular , Espectroscopia de Ressonância Magnética , Estrutura Molecular
2.
J Biol Chem ; 299(6): 104813, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37172726

RESUMO

The calmodulin-activated α-kinase, eukaryotic elongation factor 2 kinase (eEF-2K), serves as a master regulator of translational elongation by specifically phosphorylating and reducing the ribosome affinity of the guanosine triphosphatase, eukaryotic elongation factor 2 (eEF-2). Given its critical role in a fundamental cellular process, dysregulation of eEF-2K has been implicated in several human diseases, including those of the cardiovascular system, chronic neuropathies, and many cancers, making it a critical pharmacological target. In the absence of high-resolution structural information, high-throughput screening efforts have yielded small-molecule candidates that show promise as eEF-2K antagonists. Principal among these is the ATP-competitive pyrido-pyrimidinedione inhibitor, A-484954, which shows high specificity toward eEF-2K relative to a panel of "typical" protein kinases. A-484954 has been shown to have some degree of efficacy in animal models of several disease states. It has also been widely deployed as a reagent in eEF-2K-specific biochemical and cell-biological studies. However, given the absence of structural information, the precise mechanism of the A-484954-mediated inhibition of eEF-2K has remained obscure. Leveraging our identification of the calmodulin-activatable catalytic core of eEF-2K, and our recent determination of its long-elusive structure, here we present the structural basis for its specific inhibition by A-484954. This structure, which represents the first for an inhibitor-bound catalytic domain of a member of the α-kinase family, enables rationalization of the existing structure-activity relationship data for A-484954 variants and lays the groundwork for further optimization of this scaffold to attain enhanced specificity/potency against eEF-2K.


Assuntos
Trifosfato de Adenosina , Calmodulina , Quinase do Fator 2 de Elongação , Animais , Humanos , Trifosfato de Adenosina/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Quinase do Fator 2 de Elongação/antagonistas & inibidores , Quinase do Fator 2 de Elongação/química , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Fator 2 de Elongação de Peptídeos/química , Fator 2 de Elongação de Peptídeos/metabolismo , Fosforilação , Domínio Catalítico , Relação Estrutura-Atividade , Elongação Traducional da Cadeia Peptídica
3.
J Membr Biol ; 256(2): 159-174, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36454258

RESUMO

The plasma membrane and autoinhibited Ca2+-ATPases contribute to the Ca2+ homeostasis in a wide variety of organisms. The enzymatic activity of these pumps is stimulated by calmodulin, which interacts with the target protein through the calmodulin-binding domain (CaMBD). Most information about this region is related to all calmodulin modulated proteins, which indicates general chemical properties and there is no established relation between Ca2+ pump sequences and taxonomic classification. Thus, the aim of this study was to perform an in silico analysis of the CaMBD from several Ca2+-ATPases, in order to determine their diversity and to detect specific patterns and amino acid selection in different species. Patterns related to potential and confirmed CaMBD were detected using sequences retrieved from the literature. The occurrence of these patterns was determined across 120 sequences from 17 taxonomical classes, which were analyzed by a phylogenetic tree to establish phylogenetic groups. Predicted physicochemical characteristics including hydropathy and net charge were calculated for each group of sequences. 22 Ca2+-ATPases sequences from animals, unicellular eukaryotes, and plants were retrieved from bioinformatic databases. These sequences allow us to establish the Patterns 1(GQILWVRGLTRLQTQ), 3(KNPSLEALQRW), and 4(SRWRRLQAEHVKK), which are present at the beginning of putative CaMBD of metazoan, parasites, and land plants. A pattern 2 (IRVVNAFR) was consistently found at the end of most analyzed sequences. The amino acid preference in the CaMBDs changed depending on the phylogenetic groups, with predominance of several aliphatic and charged residues, to confer amphiphilic properties. The results here displayed show a conserved mechanism to contribute to the Ca2+ homeostasis across evolution and may help to detect putative CaMBDs.


Assuntos
Adenosina Trifosfatases , Calmodulina , Animais , Calmodulina/genética , Calmodulina/química , Calmodulina/metabolismo , Adenosina Trifosfatases/metabolismo , Filogenia , Membrana Celular/metabolismo , Aminoácidos/metabolismo
4.
Molecules ; 27(21)2022 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-36363988

RESUMO

In the present study, we reported the interactions at the molecular level of a series of compounds called Bisindolylmaleimide, as potential inhibitors of the calmodulin protein. Bisindolylmaleimide compounds are drug prototypes derived from Staurosporine, an alkaloid with activity for cancer treatment. Bisindolylmaleimide compounds II, IV, VII, X, and XI, are proposed and reported as possible inhibitors of calmodulin protein for the first time. For the above, a biotechnological device was used (fluorescent biosensor hCaM M124C-mBBr) to directly determine binding parameters experimentally (Kd and stoichiometry) of these compounds, and molecular modeling tools (Docking, Molecular Dynamics, and Chemoinformatic Analysis) to carry out the theoretical studies and complement the experimental data. The results indicate that this compound binds to calmodulin with a Kd between 193-248 nM, an order of magnitude lower than most classic inhibitors. On the other hand, the theoretical studies support the experimental results, obtaining an acceptable correlation between the ΔGExperimental and ΔGTheoretical (r2 = 0.703) and providing us with complementary molecular details of the interaction between the calmodulin protein and the Bisindolylmaleimide series. Chemoinformatic analyzes bring certainty to Bisindolylmaleimide compounds to address clinical steps in drug development. Thus, these results make these compounds attractive to be considered as possible prototypes of new calmodulin protein inhibitors.


Assuntos
Biofilmes , Calmodulina , Calmodulina/química , Ligantes , Reatores Biológicos , Simulação de Dinâmica Molecular , Ligação Proteica
5.
J Chem Phys ; 157(3): 035101, 2022 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-35868942

RESUMO

The mechanochemical coupling and biological function of myosin motors are regulated by Ca2+ concentrations. As one of the regulation pathways, Ca2+ binding induces a conformational change of the light chain calmodulin and its binding modes with a myosin lever arm, which can affect the stiffness of the lever arm and force transmission. However, the underlying molecular mechanism of the Ca2+ regulated stiffness change is not fully understood. Here, we study the effect of Ca2+ binding on the conformational dynamics and stiffness of the myosin VIIa lever arm bound with a calmodulin by performing molecular dynamics simulations and dynamic correlation network analysis. The results showed that the calmodulin bound lever arm at an apo state can sample three different conformations. In addition to the conformation observed in a crystal structure, a calmodulin bound lever arm at the apo condition can also adopt other two conformations featured by different extents of small-angle bending of the lever arm. However, large-angle bending is strongly prohibited. Such results suggest that the calmodulin bound lever arm without Ca2+ binding is plastic for small-angle deformation but shows high stiffness for large-angle deformation. In comparison, after the binding of Ca2+, although the calmodulin bound lever arm is locally more rigid, it can adopt largely deformed or even unfolded conformations, which may render the lever arm incompetent for force transmission. The conformational plasticity of the lever arm for small-angle deformation at the apo condition may be used as a force buffer to prevent the lever arm from unfolding during the power stroke action of the motor domain.


Assuntos
Calmodulina , Miosinas , Calmodulina/química , Conformação Molecular , Miosinas/química , Conformação Proteica
6.
Biochemistry ; 61(7): 554-562, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-35263092

RESUMO

Adenylate cyclase toxin (ACT) is a virulence factor secreted by Bordetella pertussis and plays a causative role in whooping cough. After ACT attaches to lung phagocytes, the adenylate cyclase (AC) domain of the toxin is transported into the cytoplasm where it is activated by calmodulin (CaM) to cyclize ATP into 3',5'-cyclic adenosine monophosphate (cAMP). Production of high concentrations of cAMP disrupts immune functions of phagocytes. To better understand the mechanism of activation of AC by CaM, the studies reported herein were conducted. Major observations are as follows: (1) dependence of steady-state velocities on CaM and ATP concentrations suggests that CaM and ATP bind to AC in a random fashion. (2) A pre-steady-state lag phase is observed when AC is added to solutions of CaM and ATP, reflecting the association of AC and CaM. Analysis of pre-steady-state data indicates that CaM binds to AC and AC:ATP with second-order rate constants of 30 and 60 µM-1 s-1, respectively, and that CaM dissociates from the resultant complexes with a first-order rate constant of 0.002 s-1. (3) A biphasic dependence of steady-state velocities on CaM concentration is observed: the first phase extending from 0.01 to 1 nM CaM (Kd,obs ∼ 0.06 nM) and the second phase from 1 to 2000 nM CaM (Kd,obs ∼ 60 nM). These results suggest that AC exists in at least two conformations, with each conformation exhibiting distinct binding affinity for CaM and distinct potential for activation.


Assuntos
Adenilil Ciclases , Bordetella pertussis , Toxina Adenilato Ciclase/química , Adenilil Ciclases/metabolismo , Bordetella pertussis/metabolismo , Calmodulina/química , AMP Cíclico/metabolismo , Cinética
7.
Biomolecules ; 11(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34572507

RESUMO

The HIV-1 Gag polyprotein plays essential roles during the late stage of the HIV-1 replication cycle, and has recently been identified as a promising therapeutic target. The N-terminal portion of the HIV-1 Gag polyprotein encodes the myristoylated matrix (MA) protein, which functions in the trafficking of the structural proteins to the plasma membrane (PM) and facilitation of envelope incorporation into budding virus. Numerous host cell proteins interact with the MA portion of the Gag polyprotein during this process. One such factor is the ubiquitous calcium-binding protein calmodulin (CaM), which interacts preferentially with myristoylated proteins, thereby regulating cell physiology. The exact role of this interaction is poorly understood to date. Atomic resolution structures revealed the nature of the CaM-MA interaction for clade B isolates. In this study, we expanded our knowledge and characterized biophysically and computationally the CaM interaction with MA from other HIV-1 clades and discovered differences in the CaM recognition as compared to the prototypical clade B MA, with significant alterations in the interaction with the MA protein from clade C. Structural investigation and in silico mutational analysis revealed that HIV-1 MA protein from clade C, which is responsible for the majority of global HIV-1 infections, interacts with lower affinity and altered kinetics as compared to the canonical clade B. This finding may have implications for additional altered interaction networks as compared to the well-studied clade B. Our analysis highlights the importance of expanding investigations of virus-host cell factor interaction networks to other HIV-1 clades.


Assuntos
Calmodulina/metabolismo , HIV-1/metabolismo , Proteínas da Matriz Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Calmodulina/química , Sequência Conservada , Humanos , Modelos Moleculares , Ácido Mirístico/metabolismo , Ligação Proteica , Eletricidade Estática , Proteínas da Matriz Viral/química
8.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34480001

RESUMO

RalA is a small GTPase and a member of the Ras family. This molecular switch is activated downstream of Ras and is widely implicated in tumor formation and growth. Previous work has shown that the ubiquitous Ca2+-sensor calmodulin (CaM) binds to small GTPases such as RalA and K-Ras4B, but a lack of structural information has obscured the functional consequences of these interactions. Here, we have investigated the binding of CaM to RalA and found that CaM interacts exclusively with the C terminus of RalA, which is lipidated with a prenyl group in vivo to aid membrane attachment. Biophysical and structural analyses show that the two RalA membrane-targeting motifs (the prenyl anchor and the polybasic motif) are engaged by distinct lobes of CaM and that CaM binding leads to removal of RalA from its membrane environment. The structure of this complex, along with a biophysical investigation into membrane removal, provides a framework with which to understand how CaM regulates the function of RalA and sheds light on the interaction of CaM with other small GTPases, including K-Ras4B.


Assuntos
Calmodulina/metabolismo , Bicamadas Lipídicas/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Calmodulina/química , Membrana Celular/metabolismo , Humanos , Bicamadas Lipídicas/química , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Ligação Proteica , Prenilação de Proteína , Serina/metabolismo , Proteínas ral de Ligação ao GTP/química
9.
Mol Cell ; 81(21): 4527-4539.e8, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34407442

RESUMO

The kinase domain transfers phosphate from ATP to substrates. However, the Legionella effector SidJ adopts a kinase fold, yet catalyzes calmodulin (CaM)-dependent glutamylation to inactivate the SidE ubiquitin ligases. The structural and mechanistic basis in which the kinase domain catalyzes protein glutamylation is unknown. Here we present cryo-EM reconstructions of SidJ:CaM:SidE reaction intermediate complexes. We show that the kinase-like active site of SidJ adenylates an active-site Glu in SidE, resulting in the formation of a stable reaction intermediate complex. An insertion in the catalytic loop of the kinase domain positions the donor Glu near the acyl-adenylate for peptide bond formation. Our structural analysis led us to discover that the SidJ paralog SdjA is a glutamylase that differentially regulates the SidE ligases during Legionella infection. Our results uncover the structural and mechanistic basis in which the kinase fold catalyzes non-ribosomal amino acid ligations and reveal an unappreciated level of SidE-family regulation.


Assuntos
Proteínas de Bactérias/química , Dobramento de Proteína , Proteínas/química , Fatores de Virulência/química , Proteínas de Bactérias/metabolismo , Calmodulina/química , Catálise , Domínio Catalítico , Microscopia Crioeletrônica , Legionella/enzimologia , Mutagênese , Peptídeos/química , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Espectrometria de Fluorescência , Ubiquitina-Proteína Ligases/química , Fatores de Virulência/metabolismo
10.
Nat Commun ; 12(1): 807, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33547325

RESUMO

Ryanodine Receptors (RyRs) are massive channels that release Ca2+ from the endoplasmic and sarcoplasmic reticulum. Hundreds of mutations are linked to malignant hyperthermia (MH), myopathies, and arrhythmias. Here, we explore the first MH mutation identified in humans by providing cryo-EM snapshots of the pig homolog, R615C, showing that it affects an interface between three solenoid regions. We also show the impact of apo-calmodulin (apoCaM) and how it can induce opening by bending of the bridging solenoid, mediated by its N-terminal lobe. For R615C RyR1, apoCaM binding abolishes a pathological 'intermediate' conformation, distributing the population to a mixture of open and closed channels, both different from the structure without apoCaM. Comparisons show that the mutation primarily affects the closed state, inducing partial movements linked to channel activation. This shows that disease mutations can cause distinct pathological conformations of the RyR and facilitate channel opening by disrupting interactions between different solenoid regions.


Assuntos
Apoproteínas/química , Cálcio/química , Calmodulina/química , Hipertermia Maligna/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Substituição de Aminoácidos , Animais , Apoproteínas/genética , Apoproteínas/metabolismo , Arginina/química , Arginina/metabolismo , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Microscopia Crioeletrônica , Cisteína/química , Cisteína/metabolismo , Expressão Gênica , Humanos , Transporte de Íons , Hipertermia Maligna/genética , Hipertermia Maligna/patologia , Modelos Moleculares , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Mutação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/química , Retículo Sarcoplasmático/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Suínos
11.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326637

RESUMO

Calmodulin (CaM) is a Ca2+-sensor that regulates a wide variety of target proteins, many of which interact through short basic helical motifs bearing two hydrophobic 'anchor' residues. CaM comprises two globular lobes, each containing a pair of EF-hand Ca2+-binding motifs that form a Ca2+-induced hydrophobic pocket that binds an anchor residue. A central flexible linker allows CaM to accommodate diverse targets. Several reported CaM interactors lack these anchors but contain Lys/Arg-rich polybasic sequences adjacent to a lipidated N- or C-terminus. Ca2+-CaM binds the myristoylated N-terminus of CAP23/NAP22 with intimate interactions between the lipid and a surface comprised of the hydrophobic pockets of both lobes, while the basic residues make electrostatic interactions with the negatively charged surface of CaM. Ca2+-CaM binds farnesylcysteine, derived from the farnesylated polybasic C-terminus of KRAS4b, with the lipid inserted into the C-terminal lobe hydrophobic pocket. CaM sequestration of the KRAS4b farnesyl moiety disrupts KRAS4b membrane association and downstream signaling. Phosphorylation of basic regions of N-/C-terminal lipidated CaM targets can reduce affinity for both CaM and the membrane. Since both N-terminal myristoylated and C-terminal prenylated proteins use a Singly Lipidated Polybasic Terminus (SLIPT) for CaM binding, we propose these polybasic lipopeptide elements comprise a non-canonical CaM-binding motif.


Assuntos
Sinalização do Cálcio/genética , Calmodulina/química , Calmodulina/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Motivos de Aminoácidos , Cálcio/metabolismo , Calmodulina/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Fosforilação , Plantas/química , Plantas/genética , Plantas/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Conformação Proteica , Isoformas de Proteínas , Prenilação de Proteína , Proteínas Proto-Oncogênicas p21(ras)/química , Proteínas Proto-Oncogênicas p21(ras)/genética , Eletricidade Estática
12.
Biomolecules ; 10(3)2020 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-32245175

RESUMO

Transient receptor potential (TRP) ion channels are polymodal receptors that have been implicated in a variety of pathophysiologies, including pain, obesity, and cancer. The capsaicin and heat sensor TRPV1, and the menthol and cold sensor TRPM8, have been shown to be modulated by the membrane protein PIRT (Phosphoinositide-interacting regulator of TRP). The emerging mechanism of PIRT-dependent TRPM8 regulation involves a competitive interaction between PIRT and TRPM8 for the activating phosphatidylinositol 4,5-bisphosphate (PIP2) lipid. As many PIP2 modulated ion channels also interact with calmodulin, we investigated the possible interaction between PIRT and calmodulin. Using microscale thermophoresis (MST), we show that calmodulin binds to the PIRT C-terminal α-helix, which we corroborate with a pull-down experiment, nuclear magnetic resonance-detected binding study, and Rosetta-based computational studies. Furthermore, we identify a cholesterol-recognition amino acid consensus (CRAC) domain in the outer leaflet of the first transmembrane helix of PIRT, and with MST, show that PIRT specifically binds to a number of cholesterol-derivatives. Additional studies identified that PIRT binds to cholecalciferol and oxytocin, which has mechanistic implications for the role of PIRT regulation of additional ion channels. This is the first study to show that PIRT specifically binds to a variety of ligands beyond TRP channels and PIP2.


Assuntos
Calmodulina/química , Colesterol/química , Proteínas de Membrana/química , Canais de Cátion TRPM/química , Canais de Cátion TRPV/química , Calmodulina/metabolismo , Colesterol/metabolismo , Humanos , Ligantes , Proteínas de Membrana/metabolismo , Ligação Proteica , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo
13.
Sci Signal ; 13(625)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234958

RESUMO

KRAS4b is a small guanosine triphosphatase (GTPase) protein that regulates several signal transduction pathways that underlie cell proliferation, differentiation, and survival. KRAS4b function requires prenylation of its C terminus and recruitment to the plasma membrane, where KRAS4b activates effector proteins including the RAF family of kinases. The Ca2+-sensing protein calmodulin (CaM) has been suggested to regulate the localization of KRAS4b through direct, Ca2+-dependent interaction, but how CaM and KRAS4b functionally interact is controversial. Here, we determined a crystal structure, which was supported by solution nuclear magnetic resonance (NMR), that revealed the sequestration of the prenyl moiety of KRAS4b in the hydrophobic pocket of the C-terminal lobe of Ca2+-bound CaM. Our engineered fluorescence resonance energy transfer (FRET)-based biosensor probes (CaMeRAS) showed that, upon stimulation of Ca2+ influx by extracellular ligands, KRAS4b reversibly translocated in a Ca2+-CaM-dependent manner from the plasma membrane to the cytoplasm in live HeLa and HEK293 cells. These results reveal a mechanism underlying the inhibition of KRAS4b activity by Ca2+ signaling pathways.


Assuntos
Calmodulina , Membrana Celular , Lipídeos de Membrana , Proteínas Proto-Oncogênicas p21(ras) , Calmodulina/química , Calmodulina/genética , Calmodulina/metabolismo , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Células HeLa , Humanos , Lipídeos de Membrana/química , Lipídeos de Membrana/genética , Lipídeos de Membrana/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/química , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
14.
Chemphyschem ; 21(7): 589-593, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-31755204

RESUMO

Reactions catalyzed by artificial allosteric enzymes, chimeric proteins with fused biorecognition and catalytic units, were used to mimic multi-input Boolean logic systems. The catalytic parts of the systems were represented by pyrroloquinoline quinone-dependent glucose dehydrogenase (PQQ-GDH). Two biorecognition units, calmodulin or artificial peptide-clamp, were integrated into PQQ-GDH and locked it in the OFF or ON state respectively. The ligand-peptide binding cooperatively with Ca2+ cations to a calmodulin bioreceptor resulted in the enzyme activation, while another ligand-peptide bound to a clamp-receptor inhibited the enzyme. The enzyme activation and inhibition originated from peptide-induced allosteric transitions in the receptor units that propagated to the catalytic domain. While most of enzymes used to mimic Boolean logic gates operate with two inputs (substrate and co-substrate), the used chimeric enzymes were controlled by four inputs (glucose - substrate, dichlorophenolindophenol - electron acceptor/co-substrate, Ca2+ cations and a peptide - activating/inhibiting signals). The biocatalytic reactions controlled by four input signals were considered as logic networks composed of several concatenated logic gates. The developed approach allows potentially programming complex logic networks operating with various biomolecular inputs representing potential utility for different biomedical applications.


Assuntos
Calmodulina/farmacologia , Biologia Computacional , Glucose Desidrogenase/antagonistas & inibidores , Peptídeos/farmacologia , Biocatálise , Calmodulina/química , Glucose Desidrogenase/química , Glucose Desidrogenase/metabolismo , Ligantes , Lógica , Modelos Moleculares , Estrutura Molecular , Peptídeos/química
15.
J Biomol Tech ; 30(4): 50-57, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31662705

RESUMO

Methionine oxidation plays a critical role in many processes of biologic and biomedical importance, including cellular redox responses and stability of protein pharmaceuticals. Bottom-up methods for analysis of methionine oxidation can suffer from incomplete sequence coverage, as well as an inability to readily detect correlated oxidation between 2 or more methionines. However, the methodology for quantifying protein oxidation in top-down analyses is lacking. Previous work has shown that electron transfer dissociation (ETD)-based tandem mass spectrometry (MS/MS) fragmentation offers accurate and precise quantification of amino acid oxidation in peptides, even in complex samples. However, the ability of ETD-based MS/MS fragmentation to accurately quantify amino acid oxidation of proteins in a top-down manner has not been reported. Using apomyoglobin and calmodulin as model proteins, we partially converted methionines into methionine sulfoxide by incubation in H2O2. Using top-down ETD-based fragmentation, we quantified the amount of oxidation of various ETD product ions and compared the quantified values with those from traditional bottom-up analysis. We find that overall quantification of methionine oxidation by top-down MS/MS ranges from good agreement with traditional bottom-up methods to vast differences between the 2 techniques, including missing oxidized product ions and large differences in measured oxidation quantities. Care must be taken in transitioning ETD-based quantitation of oxidation from the peptide level to the intact protein level.


Assuntos
Metionina/metabolismo , Peptídeos/química , Proteínas/química , Espectrometria de Massas em Tandem/métodos , Apoproteínas/análise , Apoproteínas/química , Calmodulina/análise , Calmodulina/química , Peróxido de Hidrogênio/química , Radical Hidroxila/química , Metionina/análise , Metionina/química , Mioglobina/análise , Mioglobina/química , Oxirredução , Peptídeos/análise , Pegadas de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas/análise , Reprodutibilidade dos Testes
16.
Curr Protein Pept Sci ; 20(11): 1102-1111, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31553290

RESUMO

Calmodulin (CaM) is a highly conserved eukaryotic Ca2+ sensor protein that is able to bind a large variety of target sequences without a defined consensus sequence. The recognition of this diverse target set allows CaM to take part in the regulation of several vital cell functions. To fully understand the structural basis of the regulation functions of CaM, the investigation of complexes of CaM and its targets is essential. In this minireview we give an outline of the different types of CaM - peptide complexes with 3D structure determined, also providing an overview of recently determined structures. We discuss factors defining the orientations of peptides within the complexes, as well as roles of anchoring residues. The emphasis is on complexes where multiple binding modes were found.


Assuntos
Calmodulina/química , Calmodulina/metabolismo , Peptídeos/metabolismo , Animais , Apoproteínas/química , Apoproteínas/metabolismo , Cálcio/metabolismo , Humanos , Ligação Proteica
17.
Structure ; 27(11): 1647-1659.e4, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31495533

RESUMO

Ca2+-calmodulin (CaM) extracts KRas4B from the plasma membrane, suggesting that KRas4B/CaM interaction plays a role in regulating Ras signaling. To gain mechanistic insight, we provide a computational model, supported by experimental structural data, of farnesylated/methylated KRas4B1-185 interacting with CaM in solution and at anionic membranes including signaling lipids. Due to multiple interaction modes, we observe diverse conformational ensembles of the KRas4B-CaM complex. A highly populated conformation reveals the catalytic domain interacting with the N-lobe and the hypervariable region (HVR) wrapping around the linker with the farnesyl docking to the extended CaM's C-lobe pocket. Alternatively, KRas4B can interact with collapsed CaM with the farnesyl penetrating CaM's center. At anionic membranes, CaM interacts with the catalytic domain with large fluctuations, drawing the HVR. Signaling lipids establishing strong salt bridges with CaM prevent membrane departure. Membrane-interacting KRas4B-CaM complex can productively recruit phosphatidylinositol 3-kinase α (PI3Kα) to the plasma membrane, serving as a coagent in activating PI3Kα/Akt signaling.


Assuntos
Calmodulina/química , Proteínas Proto-Oncogênicas p21(ras)/química , Animais , Sítios de Ligação , Calmodulina/metabolismo , Galinhas , Metilação , Simulação de Acoplamento Molecular , Prenilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
18.
Methods Enzymol ; 624: 249-264, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31370932

RESUMO

Over the past decade, photoswitchable molecules have been emerging as attractive tools for investigating biological processes with spatiotemporal resolution in a minimally invasive fashion. Photoswitches built on light-sensitive proteins or domains have significantly advanced neuronal and cellular studies. To install photosensitivity to general proteins and to enable high specificity for modulation, photoswitchable click amino acids (PSCaas) based on azobenzene have been developed and recently genetically incorporated into proteins via the expansion of the genetic code. PSCaas allow targeting selected sites in a protein for high specificity and are generally applicable to various proteins. In addition, PSCaas contain a click functional group, which selectively reacts with an appropriately positioned cysteine forming a photocontrollable bridge on the protein in situ. The photocontrollable bridge enables reversible modulation of the secondary structure of the spanned region and thus the function of the protein. In this chapter we describe the design and genetic encoding of PSCaa. Protocols are presented for incorporating PSCaa into a model protein calmodulin to build the bridge followed by photocontrol of calmodulin's conformation and binding function.


Assuntos
Aminoácidos/genética , Compostos Azo/química , Optogenética/métodos , Proteínas/genética , Aminoácidos/química , Animais , Calmodulina/química , Calmodulina/genética , Escherichia coli/química , Escherichia coli/genética , Código Genético , Humanos , Isomerismo , Luz , Modelos Moleculares , Processos Fotoquímicos , Proteínas/química
19.
Science ; 364(6442): 787-792, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31123136

RESUMO

Enzymes with a protein kinase fold transfer phosphate from adenosine 5'-triphosphate (ATP) to substrates in a process known as phosphorylation. Here, we show that the Legionella meta-effector SidJ adopts a protein kinase fold, yet unexpectedly catalyzes protein polyglutamylation. SidJ is activated by host-cell calmodulin to polyglutamylate the SidE family of ubiquitin (Ub) ligases. Crystal structures of the SidJ-calmodulin complex reveal a protein kinase fold that catalyzes ATP-dependent isopeptide bond formation between the amino group of free glutamate and the γ-carboxyl group of an active-site glutamate in SidE. We show that SidJ polyglutamylation of SidE, and the consequent inactivation of Ub ligase activity, is required for successful Legionella replication in a viable eukaryotic host cell.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/enzimologia , Ácido Poliglutâmico/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Fatores de Virulência/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Calmodulina/química , Calmodulina/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Células HEK293 , Humanos , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Fosforilação , Ácido Poliglutâmico/química , Ácido Poliglutâmico/genética , Domínios Proteicos/genética , Proteínas Quinases/química , Proteínas Quinases/genética , Ubiquitina-Proteína Ligases/genética , Fatores de Virulência/química , Fatores de Virulência/genética
20.
Angew Chem Int Ed Engl ; 58(22): 7284-7288, 2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-30938016

RESUMO

Proteins in living cells interact specifically or nonspecifically with an enormous number of biomolecules. To understand the behavior of proteins under intracellular crowding conditions, it is indispensable to observe their three-dimensional (3D) structures at the atomic level in a physiologically natural environment. We demonstrate the first de novo protein structure determinations in eukaryotes with the sf9 cell/baculovirus system using NMR data from living cells exclusively. The method was applied to five proteins, rat calmodulin, human HRas, human ubiquitin, T. thermophilus HB8 TTHA1718, and Streptococcus protein G B1 domain. In all cases, we could obtain structural information from well-resolved in-cell 3D nuclear Overhauser effect spectroscopy (NOESY) data, suggesting that our method can be a standard tool for protein structure determinations in living eukaryotic cells. For three proteins, we achieved well-converged 3D structures. Among these, the in-cell structure of protein G B1 domain was most accurately determined, demonstrating that a helix-loop region is tilted away from a ß-sheet compared to the conformation in diluted solution.


Assuntos
Algoritmos , Proteínas de Bactérias/química , Calmodulina/química , Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas Proto-Oncogênicas p21(ras)/química , Ubiquitina/química , Animais , Humanos , Modelos Moleculares , Conformação Proteica em Folha beta , Ratos , Streptococcus/metabolismo , Thermus thermophilus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA