Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 293(40): 15569-15580, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30131335

RESUMEN

Secretion of bacterial signaling proteins and adaptation to the host, especially during infection, are processes that are often linked in pathogenic bacteria. The human pathogen Staphylococcus aureus is equipped with a large arsenal of immune-modulating factors, allowing it to either subvert the host immune response or to create permissive niches for its survival. Recently, we showed that one of the low-molecular-weight protein tyrosine phosphatases produced by S. aureus, PtpA, is secreted during growth. Here, we report that deletion of ptpA in S. aureus affects intramacrophage survival and infectivity. We also observed that PtpA is secreted during macrophage infection. Immunoprecipitation assays identified several host proteins as putative intracellular binding partners for PtpA, including coronin-1A, a cytoskeleton-associated protein that is implicated in a variety of cellular processes. Of note, we demonstrated that coronin-1A is phosphorylated on tyrosine residues upon S. aureus infection and that its phosphorylation profile is linked to PtpA expression. Our results confirm that PtpA has a critical role during infection as a bacterial effector protein that counteracts host defenses.


Asunto(s)
Proteínas Bacterianas/genética , Interacciones Huésped-Patógeno , Proteínas de Microfilamentos/genética , Proteínas Tirosina Fosfatasas/genética , Infecciones Estafilocócicas/genética , Staphylococcus aureus/genética , Animales , Proteínas Bacterianas/metabolismo , Clonación Molecular , Dictyostelium/genética , Dictyostelium/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Fosforilación , Unión Proteica , Proteínas Tirosina Fosfatasas/metabolismo , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/enzimología , Staphylococcus aureus/patogenicidad , Tirosina/metabolismo , Virulencia
2.
Nat Microbiol ; 3(8): 948-959, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013238

RESUMEN

Dynamic control of cell polarity is of critical importance for many aspects of cellular development and motility. In Myxococcus xanthus, MglA, a G protein, and MglB, its cognate GTPase-activating protein, establish a polarity axis that defines the direction of movement of the cell and that can be rapidly inverted by the Frz chemosensory system. Although vital for collective cell behaviours, how Frz triggers this switch has remained unknown. Here, we use genetics, imaging and mathematical modelling to show that Frz controls polarity reversals via a gated relaxation oscillator. FrzX, which we identify as a target of the Frz kinase, provides the gating and thus acts as the trigger for reversals. Slow relocalization of the polarity protein RomR then creates a refractory period during which another switch cannot be triggered. A secondary Frz output, FrzZ, decreases this delay, allowing rapid reversals when required. Thus, this architecture results in a highly tuneable switch that allows a wide range of reversal frequencies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Myxococcus xanthus/fisiología , Polaridad Celular , Proteínas Activadoras de GTPasa/metabolismo , Modelos Teóricos , Transducción de Señal
3.
Biochem Biophys Res Commun ; 469(3): 619-25, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26679607

RESUMEN

Due to the emergence of methicillin-resistant strains, Staphylococcus aureus has become as major public-health threat. Studies aimed at deciphering the molecular mechanism of virulence are thus required to identify new targets and develop efficient therapeutic agents. Protein phosphorylations are known to play key regulatory functions and their roles in pathogenesis are under intense scrutiny. Here we analyzed the protein tyrosine phosphatase PtpA of S. aureus, a member of the family of low molecular weight protein tyrosine phosphatases that are often secreted by pathogenic bacteria. We report for the first time that PtpA is phosphorylated in vitro by the S. aureus tyrosine kinase CapA1B2. A mass spectrometry approach allowed determining that Tyr122 and Tyr123 were the only two residues phosphorylated by this kinase. This result was confirmed by analysis of a double PtpA_Y122A/Y123A mutant that showed no phosphorylation by CapA1B2. Interestingly, PtpA phosphatase activity was abrogated in this mutant, suggesting a key regulatory function for these two tyrosine residues. This was further reinforced by the observation that CapA1B2-mediated phosphorylation significantly increased PtpA phosphatase activity. Moreover, we provide evidence that PtpA is secreted during growth of S. aureus. Together our results suggest that PtpA is an exported S. aureus signaling molecule controlled by tyrosine phosphorylation which may interfere with host cell signaling.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Staphylococcus aureus/enzimología , Proliferación Celular/fisiología , Fosforilación/fisiología
4.
PLoS Genet ; 11(8): e1005460, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26291327

RESUMEN

Understanding the principles underlying the plasticity of signal transduction networks is fundamental to decipher the functioning of living cells. In Myxococcus xanthus, a particular chemosensory system (Frz) coordinates the activity of two separate motility systems (the A- and S-motility systems), promoting multicellular development. This unusual structure asks how signal is transduced in a branched signal transduction pathway. Using combined evolution-guided and single cell approaches, we successfully uncoupled the regulations and showed that the A-motility regulation system branched-off an existing signaling system that initially only controlled S-motility. Pathway branching emerged in part following a gene duplication event and changes in the circuit structure increasing the signaling efficiency. In the evolved pathway, the Frz histidine kinase generates a steep biphasic response to increasing external stimulations, which is essential for signal partitioning to the motility systems. We further show that this behavior results from the action of two accessory response regulator proteins that act independently to filter and amplify signals from the upstream kinase. Thus, signal amplification loops may underlie the emergence of new connectivity in signal transduction pathways.


Asunto(s)
Myxococcus xanthus/fisiología , Transducción de Señal , Proteínas Bacterianas/metabolismo , Quimiotaxis , Evolución Molecular , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa , Proteínas Quinasas/fisiología
5.
Cell Rep ; 12(7): 1205-16, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26257181

RESUMEN

The Piwi-interacting RNA (piRNA) pathway plays an essential role in the repression of transposons in the germline. Other functions of piRNAs such as post-transcriptional regulation of mRNAs are now emerging. Here, we perform iCLIP with the PIWI protein Aubergine (Aub) and identify hundreds of maternal mRNAs interacting with Aub in the early Drosophila embryo. Gene expression profiling reveals that a proportion of these mRNAs undergo Aub-dependent destabilization during the maternal-to-zygotic transition. Strikingly, Aub-dependent unstable mRNAs encode germ cell determinants. iCLIP with an Aub mutant that is unable to bind piRNAs confirms piRNA-dependent binding of Aub to mRNAs. Base pairing between piRNAs and mRNAs can induce mRNA cleavage and decay that are essential for embryonic development. These results suggest general regulation of maternal mRNAs by Aub and piRNAs, which plays a key developmental role in the embryo through decay and localization of mRNAs encoding germ cell determinants.


Asunto(s)
Proteínas de Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Factores de Iniciación de Péptidos/genética , Estabilidad del ARN , ARN Interferente Pequeño/genética , Animales , Drosophila/embriología , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Células Germinativas/citología , Factores de Iniciación de Péptidos/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
6.
PLoS One ; 10(3): e0119907, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25807382

RESUMEN

Here, we present for the first time that Mycobacterium tuberculosis ParB is phosphorylated by several mycobacterial Ser/Thr protein kinases in vitro. ParB and ParA are the key components of bacterial chromosome segregation apparatus. ParB is a cytosolic conserved protein that binds specifically to centromere-like DNA parS sequences and interacts with ParA, a weak ATPase required for its proper localization. Mass spectrometry identified the presence of ten phosphate groups, thus indicating that ParB is phosphorylated on eight threonines, Thr32, Thr41, Thr53, Thr110, Thr195, and Thr254, Thr300, Thr303 as well as on two serines, Ser5 and Ser239. The phosphorylation sites were further substituted either by alanine to prevent phosphorylation or aspartate to mimic constitutive phosphorylation. Electrophoretic mobility shift assays revealed a drastic inhibition of DNA-binding by ParB phosphomimetic mutant compared to wild type. In addition, bacterial two-hybrid experiments showed a loss of ParA-ParB interaction with the phosphomimetic mutant, indicating that phosphorylation is regulating the recruitment of the partitioning complex. Moreover, fluorescence microscopy experiments performed in the surrogate Mycobacterium smegmatis ΔparB strain revealed that in contrast to wild type Mtb ParB, which formed subpolar foci similar to M. smegmatis ParB, phoshomimetic Mtb ParB was delocalized. Thus, our findings highlight a novel regulatory role of the different isoforms of ParB representing a molecular switch in localization and functioning of partitioning protein in Mycobacterium tuberculosis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Segregación Cromosómica , Cromosomas Bacterianos/metabolismo , Mycobacterium tuberculosis/metabolismo , ADN Bacteriano , Fosforilación
7.
PLoS Negl Trop Dis ; 8(11): e3315, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25412098

RESUMEN

BACKGROUND: Mycobacterium ulcerans, the causative agent of Buruli ulcer in humans, is unique among the members of Mycobacterium genus due to the presence of the virulence determinant megaplasmid pMUM001. This plasmid encodes multiple virulence-associated genes, including mup011, which is an uncharacterized Ser/Thr protein kinase (STPK) PknQ. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we have characterized PknQ and explored its interaction with MupFHA (Mup018c), a FHA domain containing protein also encoded by pMUM001. MupFHA was found to interact with PknQ and suppress its autophosphorylation. Subsequent protein-protein docking and molecular dynamic simulation analyses showed that this interaction involves the FHA domain of MupFHA and PknQ activation loop residues Ser170 and Thr174. FHA domains are known to recognize phosphothreonine residues, and therefore, MupFHA may be acting as one of the few unusual FHA-domain having overlapping specificity. Additionally, we elucidated the PknQ-dependent regulation of MupDivIVA (Mup012c), which is a DivIVA domain containing protein encoded by pMUM001. MupDivIVA interacts with MupFHA and this interaction may also involve phospho-threonine/serine residues of MupDivIVA. CONCLUSIONS/SIGNIFICANCE: Together, these results describe novel signaling mechanisms in M. ulcerans and show a three-way regulation of PknQ, MupFHA, and MupDivIVA. FHA domains have been considered to be only pThr specific and our results indicate a novel mechanism of pSer as well as pThr interaction exhibited by MupFHA. These results signify the need of further re-evaluating the FHA domain -pThr/pSer interaction model. MupFHA may serve as the ideal candidate for structural studies on this unique class of modular enzymes.


Asunto(s)
Proteínas Bacterianas/química , Factores de Transcripción Forkhead/química , Mycobacterium ulcerans/química , Proteínas Serina-Treonina Quinasas/química , Proteínas Bacterianas/metabolismo , Biología Computacional , Factores de Transcripción Forkhead/metabolismo , Simulación de Dinámica Molecular , Mycobacterium ulcerans/enzimología , Mycobacterium ulcerans/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína
8.
J Biol Chem ; 289(43): 29701-11, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25193664

RESUMEN

Carbon metabolism and virulence determinant production are often linked in pathogenic bacteria, and several regulatory elements have been reported to mediate this linkage in Staphylococcus aureus. Previously, we described a novel protein, catabolite control protein E (CcpE) that functions as a regulator of the tricarboxylic acid cycle. Here we demonstrate that CcpE also regulates virulence determinant biosynthesis and pathogenesis. Specifically, deletion of ccpE in S. aureus strain Newman revealed that CcpE affects transcription of virulence factors such as capA, the first gene in the capsule biosynthetic operon; hla, encoding α-toxin; and psmα, encoding the phenol-soluble modulin cluster α. Electrophoretic mobility shift assays demonstrated that CcpE binds to the hla promoter. Mice challenged with S. aureus strain Newman or its isogenic ΔccpE derivative revealed increased disease severity in the ΔccpE mutant using two animal models; an acute lung infection model and a skin infection model. Complementation of the mutant with the ccpE wild-type allele restored all phenotypes, demonstrating that CcpE is negative regulator of virulence in S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Cápsulas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Familia de Multigenes , Pigmentos Biológicos/biosíntesis , ARN Bacteriano/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Transcripción Genética , Virulencia
9.
PLoS Pathog ; 10(5): e1004115, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809459

RESUMEN

Mycobacterium tuberculosis bacilli display two signature features: acid-fast staining and the capacity to induce long-term latent infections in humans. However, the mechanisms governing these two important processes remain largely unknown. Ser/Thr phosphorylation has recently emerged as an important regulatory mechanism allowing mycobacteria to adapt their cell wall structure/composition in response to their environment. Herein, we evaluated whether phosphorylation of KasB, a crucial mycolic acid biosynthetic enzyme, could modulate acid-fast staining and virulence. Tandem mass spectrometry and site-directed mutagenesis revealed that phosphorylation of KasB occurred at Thr334 and Thr336 both in vitro and in mycobacteria. Isogenic strains of M. tuberculosis with either a deletion of the kasB gene or a kasB_T334D/T336D allele, mimicking constitutive phosphorylation of KasB, were constructed by specialized linkage transduction. Biochemical and structural analyses comparing these mutants to the parental strain revealed that both mutant strains had mycolic acids that were shortened by 4-6 carbon atoms and lacked trans-cyclopropanation. Together, these results suggested that in M. tuberculosis, phosphorylation profoundly decreases the condensing activity of KasB. Structural/modeling analyses reveal that Thr334 and Thr336 are located in the vicinity of the catalytic triad, which indicates that phosphorylation of these amino acids would result in loss of enzyme activity. Importantly, the kasB_T334D/T336D phosphomimetic and deletion alleles, in contrast to the kasB_T334A/T336A phosphoablative allele, completely lost acid-fast staining. Moreover, assessing the virulence of these strains indicated that the KasB phosphomimetic mutant was attenuated in both immunodeficient and immunocompetent mice following aerosol infection. This attenuation was characterized by the absence of lung pathology. Overall, these results highlight for the first time the role of Ser/Thr kinase-dependent KasB phosphorylation in regulating the later stages of mycolic acid elongation, with important consequences in terms of acid-fast staining and pathogenicity.


Asunto(s)
3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/metabolismo , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Ácidos Micólicos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/genética , Animales , Proteínas Bacterianas/genética , Dominio Catalítico/genética , Pared Celular/metabolismo , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Técnicas Microbiológicas/métodos , Modelos Moleculares , Mycobacterium tuberculosis/genética , Ácidos Micólicos/química , Fosforilación , Coloración y Etiquetado/métodos , Tuberculosis/diagnóstico , Tuberculosis/metabolismo , Tuberculosis/microbiología , Virulencia
10.
Biochem Biophys Res Commun ; 447(1): 165-71, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24704444

RESUMEN

The Staphylococcus aureus Vancomycin-resistance-associated response regulator VraR is known as an important response regulator, member of the VraTSR three-component signal transduction system that modulates the expression of the cell wall stress stimulon in response to a number of different cell wall active antibiotics. Given its crucial role in regulating gene expression in response to antibiotic challenges, VraR must be tightly regulated. We report here for the first time in S. aureus convergence of two major signal transduction systems, serine/threonine protein kinase and two (three)-component systems. We demonstrate that VraR can be phosphorylated by the staphylococcal Ser/Thr protein kinase Stk1 and that phosphorylation negatively affects its DNA-binding properties. Mass spectrometric analyses and site-directed mutagenesis identified Thr106, Thr119, Thr175 and Thr178 as phosphoacceptors. A S. aureus ΔvraR mutant expressing a VraR derivative that mimics constitutive phosphorylation, VraR_Asp, still exhibited markedly decreased antibiotic resistance against different cell wall active antibiotics, when compared to the wild-type, suggesting that VraR phosphorylation may represent a novel and presumably more general mechanism of regulation of the two (three)-component systems in staphylococci.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Staphylococcus aureus/genética , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica , Pruebas de Sensibilidad Microbiana , Fosforilación , Vancomicina/metabolismo , Vancomicina/farmacología
11.
PLoS Genet ; 10(2): e1004081, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24516396

RESUMEN

Neurotrophins and their receptors control a number of cellular processes, such as survival, gene expression and axonal growth, by activating multiple signalling pathways in peripheral neurons. Whether each of these pathways controls a distinct developmental process remains unknown. Here we describe a novel knock-in mouse model expressing a chimeric TrkA/TrkC (TrkAC) receptor from TrkA locus. In these mice, prospective nociceptors survived, segregated into appropriate peptidergic and nonpeptidergic subsets, projected normally to distinct laminae of the dorsal spinal cord, but displayed aberrant peripheral target innervation. This study provides the first in vivo evidence that intracellular parts of different Trk receptors are interchangeable to promote survival and maturation of nociceptors and shows that these developmental processes can be uncoupled from peripheral target innervation. Moreover, adult homozygous TrkAC knock-in mice displayed severe deficits in acute and tissue injury-induced pain, representing the first viable adult Trk mouse mutant with a pain phenotype.


Asunto(s)
Dolor/genética , Receptor trkA/genética , Receptor trkC/genética , Médula Espinal/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Ratones , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Neuronas Aferentes/metabolismo , Nociceptores , Dolor/patología , Transducción de Señal/genética , Médula Espinal/metabolismo
12.
J Biol Chem ; 288(50): 36116-28, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24194525

RESUMEN

The tricarboxylic acid cycle (TCA cycle) is a central metabolic pathway that provides energy, reducing potential, and biosynthetic intermediates. In Staphylococcus aureus, TCA cycle activity is controlled by several regulators (e.g. CcpA, CodY, and RpiRc) in response to the availability of sugars, amino acids, and environmental stress. Developing a bioinformatic search for additional carbon catabolite-responsive regulators in S. aureus, we identified a LysR-type regulator, catabolite control protein E (CcpE), with homology to the Bacillus subtilis CcpC regulator. Inactivation of ccpE in S. aureus strain Newman revealed that CcpE is a positive transcriptional effector of the first two enzymes of the TCA cycle, aconitase (citB) and to a lesser extent citrate synthase (citZ). Consistent with the transcriptional data, aconitase activity dramatically decreased in the ccpE mutant relative to the wild-type strain. The effect of ccpE inactivation on citB transcription and the lesser effect on citZ transcription were also reflected in electrophoretic mobility shift assays where CcpE bound to the citB promoter but not the citZ promoter. Metabolomic studies showed that inactivation of ccpE resulted in increased intracellular concentrations of acetate, citrate, lactate, and alanine, consistent with a redirection of carbon away from the TCA cycle. Taken together, our data suggest that CcpE is a major direct positive regulator of the TCA cycle gene citB.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ciclo del Ácido Cítrico/genética , Proteínas Represoras/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Secuencia de Bases , Eliminación de Gen , Sitios Genéticos/genética , Metaboloma , Datos de Secuencia Molecular , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Staphylococcus aureus/crecimiento & desarrollo
13.
J Biol Chem ; 288(23): 16546-16556, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23609448

RESUMEN

GlgE is a maltosyltransferase involved in the biosynthesis of α-glucans that has been genetically validated as a potential therapeutic target against Mycobacterium tuberculosis. Despite also making α-glucan, the GlgC/GlgA glycogen pathway is distinct and allosterically regulated. We have used a combination of genetics and biochemistry to establish how the GlgE pathway is regulated. M. tuberculosis GlgE was phosphorylated specifically by the Ser/Thr protein kinase PknB in vitro on one serine and six threonine residues. Furthermore, GlgE was phosphorylated in vivo when expressed in Mycobacterium bovis bacillus Calmette-Guérin (BCG) but not when all seven phosphorylation sites were replaced by Ala residues. The GlgE orthologues from Mycobacterium smegmatis and Streptomyces coelicolor were phosphorylated by the corresponding PknB orthologues in vitro, implying that the phosphorylation of GlgE is widespread among actinomycetes. PknB-dependent phosphorylation of GlgE led to a 2 orders of magnitude reduction in catalytic efficiency in vitro. The activities of phosphoablative and phosphomimetic GlgE derivatives, where each phosphorylation site was substituted with either Ala or Asp residues, respectively, correlated with negative phosphoregulation. Complementation studies of a M. smegmatis glgE mutant strain with these GlgE derivatives, together with both classical and chemical forward genetics, were consistent with flux through the GlgE pathway being correlated with GlgE activity. We conclude that the GlgE pathway appears to be negatively regulated in actinomycetes through the phosphorylation of GlgE by PknB, a mechanism distinct from that known in the classical glycogen pathway. Thus, these findings open new opportunities to target the GlgE pathway therapeutically.


Asunto(s)
Antituberculosos , Proteínas Bacterianas/metabolismo , Glucosiltransferasas/metabolismo , Mycobacterium tuberculosis/enzimología , Proteínas Bacterianas/genética , Catálisis , Prueba de Complementación Genética , Glucosiltransferasas/genética , Mycobacterium bovis/enzimología , Mycobacterium bovis/genética , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/genética , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Streptomyces coelicolor/enzimología , Streptomyces coelicolor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...