Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
Cell ; 159(5): 1188-1199, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416954

RESUMEN

Glutamine is the primary metabolite of nitrogen assimilation from inorganic nitrogen sources in microorganisms and plants. The ability to monitor cellular nitrogen status is pivotal for maintaining metabolic homeostasis and sustaining growth. The present study identifies a glutamine-sensing mechanism common in the entire plant kingdom except Brassicaceae. The plastid-localized PII signaling protein controls, in a glutamine-dependent manner, the key enzyme of the ornithine synthesis pathway, N-acetyl-l-glutamate kinase (NAGK), that leads to arginine and polyamine formation. Crystal structures reveal that the plant-specific C-terminal extension of PII, which we term the Q loop, forms a low-affinity glutamine-binding site. Glutamine binding alters PII conformation, promoting interaction and activation of NAGK. The binding motif is highly conserved in plants except Brassicaceae. A functional Q loop restores glutamine sensing in a recombinant Arabidopsis thaliana PII protein, demonstrating the modular concept of the glutamine-sensing mechanism adopted by PII proteins during the evolution of plant chloroplasts.


Asunto(s)
Glutamina/metabolismo , Plantas/metabolismo , Secuencia de Aminoácidos , Chlamydomonas reinhardtii/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Fosfotransferasas (aceptor de Grupo Carboxilo)/química , Fosfotransferasas (aceptor de Grupo Carboxilo)/metabolismo , Plantas/clasificación , Alineación de Secuencia
2.
Proc Natl Acad Sci U S A ; 121(11): e2318320121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38457518

RESUMEN

Coordinated carbon and nitrogen metabolism is crucial for bacteria living in the fluctuating environments. Intracellular carbon and nitrogen homeostasis is maintained by a sophisticated network, in which the widespread signaling protein PII acts as a major regulatory hub. In cyanobacteria, PII was proposed to regulate the nitrate uptake by an ABC (ATP-binding cassette)-type nitrate transporter NrtABCD, in which the nucleotide-binding domain of NrtC is fused with a C-terminal regulatory domain (CRD). Here, we solved three cryoelectron microscopy structures of NrtBCD, bound to nitrate, ATP, and PII, respectively. Structural and biochemical analyses enable us to identify the key residues that form a hydrophobic and a hydrophilic cavity along the substrate translocation channel. The core structure of PII, but not the canonical T-loop, binds to NrtC and stabilizes the CRD, making it visible in the complex structure, narrows the substrate translocation channel in NrtB, and ultimately locks NrtBCD at an inhibited inward-facing conformation. Based on these results and previous reports, we propose a putative transport cycle driven by NrtABCD, which is allosterically inhibited by PII in response to the cellular level of 2-oxoglutarate. Our findings provide a distinct regulatory mechanism of ABC transporter via asymmetrically binding to a signaling protein.


Asunto(s)
Cianobacterias , Transportadores de Nitrato , Nitratos/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Alostérica , Microscopía por Crioelectrón , Cianobacterias/metabolismo , Adenosina Trifosfato/metabolismo , Nitrógeno/metabolismo , Carbono/metabolismo , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(8): e2205882120, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36800386

RESUMEN

The PII superfamily consists of widespread signal transduction proteins found in all domains of life. In addition to canonical PII proteins involved in C/N sensing, structurally similar PII-like proteins evolved to fulfill diverse, yet poorly understood cellular functions. In cyanobacteria, the bicarbonate transporter SbtA is co-transcribed with the conserved PII-like protein, SbtB, to augment intracellular inorganic carbon levels for efficient CO2 fixation. We identified SbtB as a sensor of various adenine nucleotides including the second messenger nucleotides cyclic AMP (cAMP) and c-di-AMP. Moreover, many SbtB proteins possess a C-terminal extension with a disulfide bridge of potential redox-regulatory function, which we call R-loop. Here, we reveal an unusual ATP/ADP apyrase (diphosphohydrolase) activity of SbtB that is controlled by the R-loop. We followed the sequence of hydrolysis reactions from ATP over ADP to AMP in crystallographic snapshots and unravel the structural mechanism by which changes of the R-loop redox state modulate apyrase activity. We further gathered evidence that this redox state is controlled by thioredoxin, suggesting that it is generally linked to cellular metabolism, which is supported by physiological alterations in site-specific mutants of the SbtB protein. Finally, we present a refined model of how SbtB regulates SbtA activity, in which both the apyrase activity and its redox regulation play a central role. This highlights SbtB as a central switch point in cyanobacterial cell physiology, integrating not only signals from the energy state (adenyl-nucleotide binding) and the carbon supply via cAMP binding but also from the day/night status reported by the C-terminal redox switch.


Asunto(s)
Apirasa , Cianobacterias , Apirasa/genética , Apirasa/metabolismo , Bicarbonatos/metabolismo , Proteínas Bacterianas/metabolismo , Carbono/metabolismo , Cianobacterias/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo
4.
Cell ; 139(6): 1049-51, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005798

RESUMEN

How do proteins efficiently and precisely shift from one conformation to another? Gardino et al. (2009) show that transient hydrogen bonds are critical to the conformational transition of the nitrogen regulatory protein NtrC between its native state and its active state.


Asunto(s)
Conformación Proteica , Proteínas/química , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Pliegue de Proteína , Proteínas/metabolismo
5.
Cell ; 139(6): 1109-18, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005804

RESUMEN

Phosphorylation is a common mechanism for activating proteins within signaling pathways. Yet, the molecular transitions between the inactive and active conformational states are poorly understood. Here we quantitatively characterize the free-energy landscape of activation of a signaling protein, nitrogen regulatory protein C (NtrC), by connecting functional protein dynamics of phosphorylation-dependent activation to protein folding and show that only a rarely populated, pre-existing active conformation is energetically stabilized by phosphorylation. Using nuclear magnetic resonance (NMR) dynamics, we test an atomic scale pathway for the complex conformational transition, inferred from molecular dynamics simulations (Lei et al., 2009). The data show that the loss of native stabilizing contacts during activation is compensated by non-native transient atomic interactions during the transition. The results unravel atomistic details of native-state protein energy landscapes by expanding the knowledge about ground states to transition landscapes.


Asunto(s)
Proteínas Bacterianas/química , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Conformación Proteica , Bacterias/química , Bacterias/metabolismo , Enlace de Hidrógeno , Resonancia Magnética Nuclear Biomolecular , Termodinámica
6.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526690

RESUMEN

Nitrogen limitation imposes a major transition in the lifestyle of nondiazotrophic cyanobacteria that is controlled by a complex interplay of regulatory factors involving the pervasive signal processor PII Immediately upon nitrogen limitation, newly fixed carbon is redirected toward glycogen synthesis. How the metabolic switch for diverting fixed carbon toward the synthesis of glycogen or of cellular building blocks is operated was so far poorly understood. Here, using the nondiazotrophic cyanobacterium Synechocystis sp. PCC 6803 as model system, we identified a novel PII interactor, the product of the sll0944 gene, which we named PirC. We show that PirC binds to and inhibits the activity of 2,3-phosphoglycerate-independent phosphoglycerate mutase (PGAM), the enzyme that deviates newly fixed CO2 toward lower glycolysis. The binding of PirC to either PII or PGAM is tuned by the metabolite 2-oxoglutarate (2-OG), which accumulates upon nitrogen starvation. In these conditions, the high levels of 2-OG dissociate the PirC-PII complex to promote PirC binding to and inhibition of PGAM. Accordingly, a PirC-deficient mutant showed strongly reduced glycogen levels upon nitrogen deprivation, whereas polyhydroxybutyrate granules were overaccumulated compared to wild-type. Metabolome analysis revealed an imbalance in 3-phosphoglycerate to pyruvate levels in the pirC mutant, confirming that PirC controls the carbon flux in cyanobacteria via mutually exclusive interaction with either PII or PGAM.


Asunto(s)
Proteínas Bacterianas/genética , Cianobacterias/genética , Proteínas PII Reguladoras del Nitrógeno/genética , Fosfoglicerato Mutasa/genética , Proteínas Bacterianas/metabolismo , Carbono/metabolismo , Cianobacterias/metabolismo , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Fosfoglicerato Mutasa/metabolismo , Synechocystis/genética , Synechocystis/metabolismo
7.
Int J Mol Sci ; 25(10)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38791467

RESUMEN

Yeast two-hybrid approaches, which are based on fusion proteins that must co-localise to the nucleus to reconstitute the transcriptional activity of GAL4, have greatly contributed to our understanding of the nitrogen interaction network of cyanobacteria, the main hubs of which are the trimeric PII and the monomeric PipX regulators. The bacterial two-hybrid system, based on the reconstitution in the E. coli cytoplasm of the adenylate cyclase of Bordetella pertussis, should provide a relatively faster and presumably more physiological assay for cyanobacterial proteins than the yeast system. Here, we used the bacterial two-hybrid system to gain additional insights into the cyanobacterial PipX interaction network while simultaneously assessing the advantages and limitations of the two most popular two-hybrid systems. A comprehensive mutational analysis of PipX and bacterial two-hybrid assays were performed to compare the outcomes between yeast and bacterial systems. We detected interactions that were previously recorded in the yeast two-hybrid system as negative, as well as a "false positive", the self-interaction of PipX, which is rather an indirect interaction that is dependent on PII homologues from the E. coli host, a result confirmed by Western blot analysis with relevant PipX variants. This is, to our knowledge, the first report of the molecular basis of a false positive in the bacterial two-hybrid system.


Asunto(s)
Proteínas Bacterianas , Cianobacterias , Proteínas PII Reguladoras del Nitrógeno , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Cianobacterias/metabolismo , Cianobacterias/genética , Escherichia coli/metabolismo , Escherichia coli/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/genética , Unión Proteica , Técnicas del Sistema de Dos Híbridos
8.
J Bacteriol ; 205(10): e0018123, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37791753

RESUMEN

A suite of molecular sensory systems enables Caulobacter to control growth, development, and reproduction in response to levels of essential elements. The bacterial enhancer-binding protein (bEBP) NtrC and its cognate sensor histidine kinase, NtrB, are key regulators of nitrogen assimilation in many bacteria, but their roles in Caulobacter metabolism and development are not well defined. Notably, Caulobacter NtrC is an unconventional bEBP that lacks the σ54-interacting loop commonly known as the GAFTGA motif. Here we show that deletion of Caulobacter crescentus ntrC slows cell growth in complex medium and that ntrB and ntrC are essential when ammonium is the sole nitrogen source due to their requirement for glutamine synthetase expression. Random transposition of a conserved IS3-family mobile genetic element frequently rescued the growth defect of ntrC mutant strains by restoring transcription of the glnBA operon, revealing a possible role for IS3 transposition in shaping the evolution of Caulobacter populations during nutrient limitation. We further identified dozens of direct NtrC-binding sites on the C. crescentus chromosome, with a large fraction located near genes involved in polysaccharide biosynthesis. The majority of binding sites align with those of the essential nucleoid-associated protein, GapR, or the cell cycle regulator, MucR1. NtrC is therefore predicted to directly impact the regulation of cell cycle and cell development. Indeed, loss of NtrC function led to elongated polar stalks and elevated synthesis of cell envelope polysaccharides. This study establishes regulatory connections between NtrC, nitrogen metabolism, polar morphogenesis, and envelope polysaccharide synthesis in Caulobacter. IMPORTANCE Bacteria balance cellular processes with the availability of nutrients in their environment. The NtrB-NtrC two-component signaling system is responsible for controlling nitrogen assimilation in many bacteria. We have characterized the effect of ntrB and ntrC deletion on Caulobacter growth and development and uncovered a role for spontaneous IS element transposition in the rescue of transcriptional and nutritional deficiencies caused by ntrC mutation. We further defined the regulon of Caulobacter NtrC, a bacterial enhancer-binding protein, and demonstrate that it shares specific binding sites with essential proteins involved in cell cycle regulation and chromosome organization. Our work provides a comprehensive view of transcriptional regulation mediated by a distinctive NtrC protein, establishing its connection to nitrogen assimilation and developmental processes in Caulobacter.


Asunto(s)
Caulobacter , Secuencia de Bases , Caulobacter/genética , Nitrógeno/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Polisacáridos , Regulación Bacteriana de la Expresión Génica , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo
9.
Mol Microbiol ; 116(4): 1216-1231, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34494331

RESUMEN

Uropathogenic Escherichia coli (UPEC) is the primary causative agent of urinary tract infections (UTIs). Successful urinary tract colonization requires appropriate expression of virulence factors in response to host environmental cues, such as limited oxygen and iron availability. Hemolysin is a pore-forming toxin, and its expression correlates with the severity of UPEC infection. Previously, we showed that hemolysin expression is enhanced under anaerobic conditions; however, the genetic basis and regulatory mechanisms involved remain undefined. Here, a transposon-based forward screen identified bis-molybdopterin guanine dinucleotide cofactor (bis-MGD) biosynthesis as an important factor for a full transcription of hemolysin under anaerobiosis but not under aerobiosis. bis-MGD positively influences hemolysin transcription via c3566-c3568, an operon immediately upstream of and cotranscribed with hlyCABD. Furthermore, suppressor mutation analysis identified the nitrogen regulator NtrC as a direct repressor of c3566-c3568-hlyCABD expression, and intact bis-MGD biosynthesis downregulated ntrC expression, thus at least partially explaining the positive role of bis-MGD in modulating hemolysin expression. Finally, bis-MGD is involved in hemolysin-mediated uroepithelial cell death and contributes to the competitive fitness of UPEC in a murine model of UTI. Collectively, our data establish that bis-MGD biosynthesis plays a crucial role in UPEC fitness in vivo, thus providing a potential target for combatting UTIs.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Nucleótidos de Guanina/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Pterinas/metabolismo , Escherichia coli Uropatógena/genética , Escherichia coli Uropatógena/metabolismo , Anaerobiosis , Animales , Muerte Celular , Línea Celular , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos CBA , Mutagénesis Insercional , Operón , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
Mol Microbiol ; 115(4): 526-538, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33012071

RESUMEN

Escherichia coli uses the C4-dicarboxylate transporter DcuA for L-aspartate/fumarate antiport, which results in the exploitation of L-aspartate for fumarate respiration under anaerobic conditions and for nitrogen assimilation under aerobic and anaerobic conditions. L-Aspartate represents a high-quality nitrogen source for assimilation. Nitrogen assimilation from L-aspartate required DcuA, and aspartase AspA to release ammonia. Ammonia is able to provide by established pathways the complete set of intracellular precursors (ammonia, L-aspartate, L-glutamate, and L-glutamine) for synthesizing amino acids, nucleotides, and amino sugars. AspA was regulated by a central regulator of nitrogen metabolism, GlnB. GlnB interacted with AspA and stimulated its L-aspartate deaminase activity (NH3 -forming), but not the reverse amination reaction. GlnB stimulation required 2-oxoglutarate and ATP, or uridylylated GlnB-UMP, consistent with the activation of nitrogen assimilation under nitrogen limitation. Binding to AspA was lost in the GlnB(Y51F) mutant of the uridylylation site. AspA, therefore, represents a new type of GlnB target that binds GlnB (with ATP and 2-oxoglutarate), or GlnB-UMP (with or without effectors), and both situations stimulate AspA deamination activity. Thus, AspA represents the central enzyme for nitrogen assimilation from L-aspartate, and AspA is integrated into the nitrogen assimilation network by the regulator GlnB.


Asunto(s)
Aspartato Amoníaco-Liasa/metabolismo , Ácido Aspártico/metabolismo , Proteínas Bacterianas/metabolismo , Transportadores de Ácidos Dicarboxílicos/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Escherichia coli/fisiología , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Amoníaco/metabolismo , Ácido Aspártico/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Redes y Vías Metabólicas , Mutación , Proteínas PII Reguladoras del Nitrógeno/química , Proteínas PII Reguladoras del Nitrógeno/genética , Dominios y Motivos de Interacción de Proteínas
11.
Mol Microbiol ; 114(2): 292-307, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32274833

RESUMEN

Phosphoenolpyruvate carboxylase (PEPC) is the second major carbon-fixing enzyme in photoautotrophic organisms. PEPC is required for the synthesis of amino acids of the glutamate and aspartate family by replenishing the TCA cycle. Furthermore, in cyanobacteria, PEPC, together with malate dehydrogenase and malic enzyme, forms a metabolic shunt for the synthesis of pyruvate from PEP. During this process, CO2 is first fixed and later released again. Due to its central metabolic position, it is crucial to fully understand the regulation of PEPC. Here, we identify PEPC from the cyanobacterium Synechocystis sp. PCC 6803 (PEPC) as a novel interaction partner for the global signal transduction protein PII . In addition to an extensive characterization of PEPC, we demonstrate specific PII -PEPC complex formation and its enzymatic consequences. PEPC activity is tuned by the metabolite-sensing properties of PII : Whereas in the absence of PII, PEPC is subjected to ATP inhibition, it is activated beyond its basal activity in the presence of PII . Furthermore, PII -PEPC complex formation is inhibited by ADP and PEPC activation by PII -ATP is mitigated in the presence of 2-OG, linking PEPC regulation to the cell's global carbon/nitrogen status. Finally, physiological relevance of the in vitro measurements was proven by metabolomic analyses of Synechocystis wild-type and PII -deficient cells.


Asunto(s)
Proteínas PII Reguladoras del Nitrógeno/metabolismo , Fosfoenolpiruvato Carboxilasa/metabolismo , Synechocystis/metabolismo , Carbono/metabolismo , Cianobacterias/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/fisiología , Fosfoenolpiruvato Carboxilasa/fisiología , Fosforilación , Transducción de Señal/fisiología , Synechocystis/fisiología
12.
Plant Cell Physiol ; 62(4): 721-731, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-33650637

RESUMEN

In cyanobacteria, the PII protein (the glnB gene product) regulates a number of proteins involved in nitrogen assimilation including PipX, the coactivator of the global nitrogen regulator protein NtcA. In Synechococcus elongatus PCC 7942, construction of a PII-less mutant retaining the wild-type pipX gene is difficult because of the toxicity of uncontrolled action of PipX and the other defect(s) resulting from the loss of PIIper se, but the nature of the PipX toxicity and the PipX-independent defect(s) remains unclear. Characterization of a PipX-less glnB mutant (PD4) in this study showed that the loss of PII increases the sensitivity of PSII to ammonium. Ammonium was shown to stimulate the formation of reactive oxygen species in the mutant cells. The ammonium-sensitive growth phenotype of PD4 was rescued by the addition of an antioxidant α-tocopherol, confirming that photo-oxidative damage was the major cause of the growth defect. A targeted PII mutant retaining wild-type pipX was successfully constructed from the wild-type S. elongatus strain (SPc) in the presence of α-tocopherol. The resulting mutant (PD1X) showed an unusual chlorophyll fluorescence profile, indicating extremely slow reduction and re-oxidation of QA, which was not observed in mutants defective in both glnB and pipX. These results showed that the aberrant action of uncontrolled PipX resulted in an impairment of the electron transport reactions in both the reducing and oxidizing sides of QA.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Synechococcus/crecimiento & desarrollo , Synechococcus/metabolismo , Compuestos de Amonio/metabolismo , Compuestos de Amonio/farmacología , Proteínas Bacterianas/genética , Clorofila/química , Clorofila/metabolismo , Medios de Cultivo/química , Medios de Cultivo/farmacología , Fluorescencia , Mutación , Proteínas PII Reguladoras del Nitrógeno/genética , Paraquat/farmacología , Especies Reactivas de Oxígeno , Synechococcus/efectos de los fármacos , Synechococcus/genética , alfa-Tocoferol/farmacología
13.
Genomics ; 112(5): 3497-3503, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32562829

RESUMEN

Nitrogen (N) element is essential nutrient, and affect metabolism of secondary metabolites in higher plants. Ascorbate peroxidase (APX) plays an important role in ascorbic acid (AsA) metabolism of tea plant. However, the roles of cytosolic ascorbate peroxidase 1 (CsAPX1) in AsA metabolism under N deficiency stress in tea plant remains unclear in detail. In this work, nitrogen regulatory protein P-II (CsGLB1) and CsAPX1 were identified by isobaric tags for relative and absolute quantitation (iTRAQ) from tea plant. The cell growth rates in transgenic Escherichia coli overexpressing CsAPX1 and CsGLB1 were higher than empty vector under N sufficiency condition. Phenotype of shoots and roots, AsA accumulation, and expression levels of AtAPX1 and AtGLB1 genes were changed in transgenic Arabidopsis hosting CsAPX1 under N deficiency stress. These findings suggested that cytosolic CsAPX1 acted a regulator in AsA accumulation through cooperating with GLB1 under N deficiency stress in tea plant.


Asunto(s)
Ascorbato Peroxidasas/metabolismo , Ácido Ascórbico/metabolismo , Camellia sinensis/metabolismo , Nitrógeno/fisiología , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Proteínas de Plantas/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Ascorbato Peroxidasas/genética , Camellia sinensis/enzimología , Camellia sinensis/genética , Escherichia coli/crecimiento & desarrollo , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas de Plantas/genética , Regiones Promotoras Genéticas , Estrés Fisiológico/genética
14.
Int J Mol Sci ; 22(23)2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34884470

RESUMEN

The PII protein is an evolutionary, highly conserved regulatory protein found in both bacteria and higher plants. In bacteria, it modulates the activity of several enzymes, transporters, and regulatory factors by interacting with them and thereby regulating important metabolic hubs, such as carbon/nitrogen homeostasis. More than two decades ago, the PII protein was characterized for the first time in plants, but its physiological role is still not sufficiently resolved. To gain more insights into the function of this protein, we investigated the interaction behavior of AtPII with candidate proteins by BiFC and FRET/FLIM in planta and with GFP/RFP traps in vitro. In the course of these studies, we found that AtPII interacts in chloroplasts with itself as well as with known interactors such as N-acetyl-L-glutamate kinase (NAGK) in dot-like aggregates, which we named PII foci. In these novel protein aggregates, AtPII also interacts with yet unknown partners, which are known to be involved in plastidic protein degradation. Further studies revealed that the C-terminal component of AtPII is crucial for the formation of PII foci. Altogether, the discovery and description of PII foci indicate a novel mode of interaction between PII proteins and other proteins in plants. These findings may represent a new starting point for the elucidation of physiological functions of PII proteins in plants.


Asunto(s)
Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Cloroplastos/metabolismo , Proteínas PII Reguladoras del Nitrógeno/química , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Dominios Proteicos , Mapas de Interacción de Proteínas , Proteolisis
15.
Int J Mol Sci ; 22(16)2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34445335

RESUMEN

Protein inhibition is a natural regulatory process to control cellular metabolic fluxes. PII-family signal-transducing effectors are in this matter key regulators of the nitrogen metabolism. Their interaction with their various targets is governed by the cellular nitrogen level and the energy charge. Structural studies on GlnK, a PII-family inhibitor of the ammonium transporters (Amt), showed that the T-loops responsible for channel obstruction are displaced upon the binding of 2-oxoglutarate, magnesium and ATP in a conserved cleft. However, GlnK from Methanocaldococcus jannaschii was shown to bind 2-oxoglutarate on the tip of its T-loop, causing a moderate disruption to GlnK-Amt interaction, raising the question if methanogenic archaea use a singular adaptive strategy. Here we show that membrane fractions of Methanothermococcus thermolithotrophicus released GlnKs only in the presence of Mg-ATP and 2-oxoglutarate. This observation led us to structurally characterize the two GlnK isoforms apo or in complex with ligands. Together, our results show that the 2-oxoglutarate binding interface is conserved in GlnKs from Methanococcales, including Methanocaldococcus jannaschii, emphasizing the importance of a free carboxy-terminal group to facilitate ligand binding and to provoke the shift of the T-loop positions.


Asunto(s)
Compuestos de Amonio/metabolismo , Ácidos Cetoglutáricos/metabolismo , Methanococcales/metabolismo , Proteínas PII Reguladoras del Nitrógeno , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Secuencia Conservada , Cristalografía por Rayos X , Transporte Iónico , Redes y Vías Metabólicas , Modelos Moleculares , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/química , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas/fisiología , Análisis de Secuencia de Proteína
16.
J Bacteriol ; 202(19)2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32690554

RESUMEN

Listeria monocytogenes is a fastidious bacterial pathogen that can utilize only a limited number of nitrogen sources for growth. Both glutamine and ammonium are common nitrogen sources used in listerial defined growth media, but little is known about the regulation of their uptake or utilization. The functional role of L. monocytogenes GlnR, the transcriptional regulator of nitrogen metabolism genes in low-G+C Gram-positive bacteria, was determined using transcriptome sequencing and real-time reverse transcription-PCR experiments. The GlnR regulon included transcriptional units involved in ammonium transport (amtB glnK) and biosynthesis of glutamine (glnRA) and glutamate (gdhA) from ammonium. As in other bacteria, GlnR proved to be an autoregulatory repressor of the glnRA operon. Unexpectedly, GlnR was most active during growth with ammonium as the nitrogen source and less active in the glutamine medium, apparently because listerial cells perceive growth with glutamine as a nitrogen-limiting condition. Therefore, paradoxically, expression of the glnA gene, encoding glutamine synthetase, was highest in the glutamine medium. For the amtB glnK operon, GlnR served as both a negative regulator in the presence of ammonium and a positive regulator in the glutamine medium. The gdhA gene was subject to a third mode of regulation that apparently required an elevated level of GlnR for repression. Finally, activity of glutamate dehydrogenase encoded by the gdhA gene appeared to correlate inversely with expression of gltAB, the operon that encodes the other major glutamate-synthesizing enzyme, glutamate synthase. Both gdhA and amtB were also regulated, in a negative manner, by the global transcriptional regulator CodY.IMPORTANCEL. monocytogenes is a widespread foodborne pathogen. Nitrogen-containing compounds, such as the glutamate-containing tripeptide, glutathione, and glutamine, have been shown to be important for expression of L. monocytogenes virulence genes. In this work, we showed that a transcriptional regulator, GlnR, controls expression of critical listerial genes of nitrogen metabolism that are involved in ammonium uptake and biosynthesis of glutamine and glutamate. A different mode of GlnR-mediated regulation was found for each of these three pathways.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Nitrógeno/metabolismo , Compuestos de Amonio/metabolismo , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/metabolismo , Ácido Glutámico/biosíntesis , Ácido Glutámico/genética , Glutamina/biosíntesis , Glutamina/genética , Listeria monocytogenes/crecimiento & desarrollo , Mutación , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Operón , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Regiones Promotoras Genéticas , RNA-Seq , Regulón , Transactivadores/genética , Transactivadores/metabolismo , Transcriptoma , Virulencia/genética
17.
J Proteome Res ; 19(1): 92-105, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31599156

RESUMEN

Azospirillum brasilense is a diazotrophic microorganism capable of associating with roots of important grasses and cereals, promoting plant growth and increasing crop yields. Nitrogen levels and the Ntr regulatory system control the nitrogen metabolism in A. brasilense. This system comprises the nitrogen regulatory proteins GlnD, which is capable of adding uridylyl groups to the PII proteins, GlnB (PII-1) and GlnZ (PII-2), under limiting nitrogen levels. Under such conditions, the histidine kinase NtrB (nitrogen regulatory protein B) cannot interact with GlnB and phosphorylate NtrC (nitrogen regulatory protein C). The phosphorylated form of NtrC acts as a transcriptional activator of genes involved in the metabolism of alternative nitrogen sources. Considering the key role of NtrC in nitrogen metabolism in A. brasilense, in this work we evaluated the proteomic and metabolomic profiles of the wild-type FP2 strain and its mutant ntrC grown under high and low nitrogen. Analysis of the integrated data identifies novel NtrC targets, including proteins involved in the response against oxidative stress (i.e., glutathione S-transferase and hydroperoxide resistance protein), underlining the importance of NtrC to bacterial survival under oxidative stress conditions.


Asunto(s)
Azospirillum brasilense , Proteómica , Azospirillum brasilense/genética , Azospirillum brasilense/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Nitrógeno/metabolismo , Fijación del Nitrógeno , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo
18.
Appl Environ Microbiol ; 86(8)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32060028

RESUMEN

PII signal transduction proteins are ubiquitous and highly conserved in bacteria, archaea, and plants and play key roles in controlling nitrogen metabolism. However, research on biological functions and regulatory targets of PII proteins remains limited. Here, we illustrated experimentally that the PII protein Corynebacterium glutamicum GlnK (CgGlnK) increased l-arginine yield when glnK was overexpressed in Corynebacterium glutamicum Data showed that CgGlnK regulated l-arginine biosynthesis by upregulating the expression of genes of the l-arginine metabolic pathway and interacting with N-acetyl-l-glutamate kinase (CgNAGK), the rate-limiting enzyme in l-arginine biosynthesis. Further assays indicated that CgGlnK contributed to alleviation of the feedback inhibition of CgNAGK caused by l-arginine. In silico analysis of the binding interface of CgGlnK-CgNAGK suggested that the B and T loops of CgGlnK mainly interacted with C and N domains of CgNAGK. Moreover, F11, R47, and K85 of CgGlnK were identified as crucial binding sites that interact with CgNAGK via hydrophobic interaction and H bonds, and these interactions probably had a positive effect on maintaining the stability of the complex. Collectively, this study reveals PII-NAGK interaction in nonphotosynthetic microorganisms and further provides insights into the regulatory mechanism of PII on amino acid biosynthesis in corynebacteria.IMPORTANCE Corynebacteria are safe industrial producers of diverse amino acids, including l-glutamic acid and l-arginine. In this study, we showed that PII protein GlnK played an important role in l-glutamic acid and l-arginine biosynthesis in C. glutamicum Through clarifying the molecular mechanism of CgGlnK in l-arginine biosynthesis, the novel interaction between CgGlnK and CgNAGK was revealed. The alleviation of l-arginine inhibition of CgNAGK reached approximately 48.21% by CgGlnK addition, and the semi-inhibition constant of CgNAGK increased 1.4-fold. Furthermore, overexpression of glnK in a high-yield l-arginine-producing strain and fermentation of the recombinant strain in a 5-liter bioreactor led to a remarkably increased production of l-arginine, 49.978 g/liter, which was about 22.61% higher than that of the initial strain. In conclusion, this study provides a new strategy for modifying amino acid biosynthesis in C. glutamicum.


Asunto(s)
Arginina/metabolismo , Proteínas Bacterianas/genética , Corynebacterium glutamicum/genética , Proteínas PII Reguladoras del Nitrógeno/genética , Fosfotransferasas (aceptor de Grupo Carboxilo)/genética , Transducción de Señal , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Corynebacterium glutamicum/química , Corynebacterium glutamicum/metabolismo , Proteínas PII Reguladoras del Nitrógeno/química , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Fosfotransferasas (aceptor de Grupo Carboxilo)/metabolismo , Alineación de Secuencia
19.
New Phytol ; 227(3): 722-731, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32077495

RESUMEN

The PII superfamily consists of signal transduction proteins found in all domains of life. Canonical PII proteins sense the cellular energy state through the competitive binding of ATP and ADP, and carbon/nitrogen balance through 2-oxoglutarate binding. The ancestor of Archaeplastida inherited its PII signal transduction protein from an ancestral cyanobacterial endosymbiont. Over the course of evolution, plant PII proteins acquired a glutamine-sensing C-terminal extension, subsequently present in all Chloroplastida PII proteins. The PII proteins of various algal strains (red, green and nonphotosynthetic algae) have been systematically investigated with respect to their sensory and regulatory properties. Comparisons of the PII proteins from different phyla of oxygenic phototrophs (cyanobacteria, red algae, Chlorophyta and higher plants) have yielded insights into their evolutionary conservation vs adaptive properties. The highly conserved role of the controlling enzyme of arginine biosynthesis, N-acetyl-l-glutamate kinase (NAGK), as a main PII-interactor has been demonstrated across oxygenic phototrophs of cyanobacteria and Archaeplastida. In addition, the PII signalling system of red algae has been identified as an evolutionary intermediate between that of Cyanobacteria and Chloroplastida. In this review, we consider recent advances in understanding metabolic signalling by PII proteins of the plant kingdom.


Asunto(s)
Cianobacterias , Rhodophyta , Proteínas Bacterianas/metabolismo , Cianobacterias/genética , Cianobacterias/metabolismo , Ácidos Cetoglutáricos , Proteínas PII Reguladoras del Nitrógeno/genética , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Transducción de Señal
20.
J Bacteriol ; 201(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30420451

RESUMEN

To cope with fluctuations in their environment, bacteria have evolved multiple adaptive stress responses. One such response is the nitrogen regulation stress response, which allows bacteria, such as Escherichia coli, to cope with and overcome conditions of nitrogen limitation. This response is directed by the two-component system NtrBC, where NtrC acts as the major transcriptional regulator to activate the expression of genes to mount the response. Recently, my colleagues and I showed that NtrC directly regulates the expression of the relA gene, the major (p)ppGpp synthetase in E. coli, coupling the nitrogen regulation stress and stringent responses. As elevated levels of (p)ppGpp have been implicated in the formation of persister cells, here, I investigated whether nitrogen starvation promotes their formation and whether the NtrC-RelA regulatory cascade plays a role. The results reveal that nitrogen-starved E. coli synthesizes (p)ppGpp and forms a higher percentage of persister cells than nonstarved cells and that both NtrC and RelA are important for these processes. This study provides novel insights into how the formation of persisters can be promoted in response to a nutritional stress.IMPORTANCE Bacteria often reside in environments where nutrient availability is scarce; therefore, they have evolved adaptive responses to rapidly cope with conditions of feast and famine. Understanding the mechanisms that underpin the regulation of how bacteria cope with this stress is a fundamentally important question in the wider context of understanding the biology of the bacterial cell and bacterial pathogenesis. Two major adaptive mechanisms to cope with starvation are the nitrogen regulation (ntr) stress and stringent responses. Here, I describe how these bacterial stress responses are coordinated under conditions of nitrogen starvation to promote the formation of antibiotic-tolerant persister cells by elevating levels of the secondary messenger (p)ppGpp.


Asunto(s)
Adaptación Fisiológica , Proteínas de Escherichia coli/metabolismo , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , GTP Pirofosfoquinasa/metabolismo , Nitrógeno/metabolismo , Proteínas PII Reguladoras del Nitrógeno/metabolismo , Estrés Fisiológico , Factores de Transcripción/metabolismo , Regulación Bacteriana de la Expresión Génica , Guanosina Pentafosfato/metabolismo , Guanosina Tetrafosfato/metabolismo , Complejos Multienzimáticos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA