Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO J ; 43(13): 2636-2660, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38778156

RESUMEN

During infection viruses hijack host cell metabolism to promote their replication. Here, analysis of metabolite alterations in macrophages exposed to poly I:C recognises that the antiviral effector Protein Kinase RNA-activated (PKR) suppresses glucose breakdown within the pentose phosphate pathway (PPP). This pathway runs parallel to central glycolysis and is critical to producing NADPH and pentose precursors for nucleotides. Changes in metabolite levels between wild-type and PKR-ablated macrophages show that PKR controls the generation of ribose 5-phosphate, in a manner distinct from its established function in gene expression but dependent on its kinase activity. PKR phosphorylates and inhibits the Ribose 5-Phosphate Isomerase A (RPIA), thereby preventing interconversion of ribulose- to ribose 5-phosphate. This activity preserves redox control but decreases production of ribose 5-phosphate for nucleotide biosynthesis. Accordingly, the PKR-mediated immune response to RNA suppresses nucleic acid production. In line, pharmacological targeting of the PPP during infection decreases the replication of the Herpes simplex virus. These results identify an immune response-mediated control of host cell metabolism and suggest targeting the RPIA as a potential innovative antiviral treatment.


Asunto(s)
Macrófagos , Vía de Pentosa Fosfato , Ribosamonofosfatos , eIF-2 Quinasa , Animales , Ribosamonofosfatos/metabolismo , Ratones , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Isomerasas Aldosa-Cetosa/metabolismo , Isomerasas Aldosa-Cetosa/genética , ARN/metabolismo , ARN/genética , Poli I-C/farmacología , Ácidos Nucleicos/metabolismo , Ácidos Nucleicos/inmunología , Replicación Viral , Fosforilación
2.
Int J Mol Sci ; 23(14)2022 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-35887232

RESUMEN

Ribose-5-phosphate isomerase A (RPIA) regulates tumorigenesis in liver and colorectal cancer. However, the role of RPIA in lung cancer remains obscure. Here we report that the suppression of RPIA diminishes cellular proliferation and activates autophagy, apoptosis, and cellular senescence in lung cancer cells. First, we detected that RPIA protein was increased in the human lung cancer versus adjust normal tissue via tissue array. Next, the knockdown of RPIA in lung cancer cells displayed autophagic vacuoles, enhanced acridine orange staining, GFP-LC3 punctae, accumulated autophagosomes, and showed elevated levels of LC3-II and reduced levels of p62, together suggesting that the suppression of RPIA stimulates autophagy in lung cancer cells. In addition, decreased RPIA expression induced apoptosis by increasing levels of Bax, cleaved PARP and caspase-3 and apoptotic cells. Moreover, RPIA knockdown triggered cellular senescence and increased p53 and p21 levels in lung cancer cells. Importantly, RPIA knockdown elevated reactive oxygen species (ROS) levels. Treatment of ROS scavenger N-acetyl-L-cysteine (NAC) reverts the activation of autophagy, apoptosis and cellular senescence by RPIA knockdown in lung cancer cells. In conclusion, RPIA knockdown induces ROS levels to activate autophagy, apoptosis, and cellular senescence in lung cancer cells. Our study sheds new light on RPIA suppression in lung cancer therapy.


Asunto(s)
Autofagia , Neoplasias Pulmonares , Isomerasas Aldosa-Cetosa , Apoptosis , Línea Celular Tumoral , Senescencia Celular , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
Antimicrob Agents Chemother ; 65(7): e0189220, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33875438

RESUMEN

Neglected tropical diseases caused by kinetoplastid parasites (Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp.) place a significant health and economic burden on developing nations worldwide. Current therapies are largely outdated, inadequate, and face mounting drug resistance from the causative parasites. Thus, there is an urgent need for drug discovery and development. Target-led drug discovery approaches have focused on the identification of parasite enzymes catalyzing essential biochemical processes, which significantly differ from equivalent proteins found in humans, thereby providing potentially exploitable therapeutic windows. One such target is ribose 5-phosphate isomerase B (RpiB), an enzyme involved in the nonoxidative branch of the pentose phosphate pathway, which catalyzes the interconversion of d-ribose 5-phosphate and d-ribulose 5-phosphate. Although protozoan RpiB has been the focus of numerous targeted studies, compounds capable of selectively inhibiting this parasite enzyme have not been identified. Here, we present the results of a fragment library screening against Leishmania infantum RpiB (LiRpiB), performed using thermal shift analysis. Hit fragments were shown to be effective inhibitors of LiRpiB in activity assays, and several fragments were capable of selectively inhibiting parasite growth in vitro. These results support the identification of LiRpiB as a validated therapeutic target. The X-ray crystal structure of apo LiRpiB was also solved, permitting docking studies to assess how hit fragments might interact with LiRpiB to inhibit its activity. Overall, this work will guide structure-based development of LiRpiB inhibitors as antileishmanial agents.


Asunto(s)
Leishmania infantum , Preparaciones Farmacéuticas , Secuencia de Aminoácidos , Humanos , Ribosamonofosfatos
4.
Appl Microbiol Biotechnol ; 105(2): 509-523, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33394147

RESUMEN

Ribose-5-phosphate isomerase B (RpiB) was first identified in the pentose phosphate pathway responsible for the inter-conversion of ribose-5-phosphate and ribulose-5-phosphate. Though there are seldom key enzymes in central carbon metabolic system developed as useful biocatalysts, RpiB with the advantages of wide substrate scope and high stereoselectivity has become a potential biotechnological tool to fulfill the demand of rare sugars currently. In this review, the pivotal roles of RpiB in carbon metabolism are summarized, and their sequence identity and structural similarity are discussed. Substrate binding and catalytic mechanisms are illustrated to provide solid foundations for enzyme engineering. Interesting differences in origin, physiological function, structure, and catalytic mechanism between RpiB and ribose-5-phosphate isomerase A are introduced. Moreover, enzyme engineering efforts for rare sugar production are stressed, and prospects of future development are concluded briefly in the viewpoint of biocatalysis. Aided by the progresses of structural and computational biology, the application of RpiB will be promoted greatly in the preparation of valuable molecules. KEY POINTS: • Detailed illustration of RpiB's vital function in central carbon metabolism. • Potential of RpiB in sequence, substrate scope, and mechanism for application. • Enzyme engineering efforts to promote RpiB in the preparation of rare sugars.


Asunto(s)
Isomerasas Aldosa-Cetosa , Azúcares , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/metabolismo , Carbono , Vía de Pentosa Fosfato
5.
Biotechnol Lett ; 43(7): 1277-1287, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33797654

RESUMEN

OBJECTIVE: Erythritol (1,2,3,4-butanetetrol) is a 4-carbon sugar alcohol that occurs in nature as a metabolite or storage compound. In this study, a multiple gene integration strategy was employed to enhance erythritol production in Y. lipolytica. RESULTS: The effects on the production of erythritol in Y. lipolytica of seven key genes involved in the erythritol synthesis pathway were evaluated individually, among which transketolase (TKL1) and transaldolase (TAL1) showed important roles in enhancing erythritol production. The combined overexpression of four genes (GUT1, TPI1, TKL1, TAL1) and disruption of the EYD1 gene (encoding erythritol dehydrogenase), resulted in produce approximately 40 g/L erythritol production from glycerol. Further enhanced erythritol synthesis was obtained by overexpressing the RKI1 gene (encoding ribose 5-phosphate isomerase) and the AMPD gene (encoding AMP deaminase), indicating for the first time that these two genes are also related to the enhancement of erythritol production in Y. lipolytica. CONCLUSIONS: A combined gene overexpression strategy was developed to efficiently improve the production of erythritol in Y. lipolytica, suggesting a great capacity and promising potential of this non-conventional yeast in converting glycerol into erythritol.


Asunto(s)
Eritritol/biosíntesis , Proteínas Fúngicas/genética , Ingeniería Metabólica/métodos , Yarrowia/crecimiento & desarrollo , AMP Desaminasa/genética , Isomerasas Aldosa-Cetosa/genética , Técnicas de Cultivo Celular por Lotes , Glicerol/metabolismo , Transaldolasa/genética , Transcetolasa/genética , Yarrowia/genética , Yarrowia/metabolismo
6.
Appl Microbiol Biotechnol ; 104(15): 6429-6441, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32533303

RESUMEN

Ribose-5-phosphate isomerase (Rpi, EC 5.3.1.6) is widespread in microorganisms, animals, and plants. It has a pivotal role in the pentose phosphate pathway and responsible for catalyzing the isomerization between D-ribulose 5-phosphate and D-ribose 5-phosphate. In recent years, Rpi has received considerable attention as a multipurpose biocatalyst for production of rare sugars, including D-allose, L-rhamnulose, L-lyxose, and L-tagatose. Besides, it has been thought of as a potential drug target in the treatment of trypanosomatid-caused diseases such as Chagas' disease, leishmaniasis, and human African trypanosomiasis. Despite increased research activities, up to now, no systematic review of Rpi has been published. To fill this gap, this paper provides detailed information about the enzymatic properties of various Rpis. Furthermore, structural features, catalytic mechanism, and molecular modifications of Rpis are summarized based on extensive crystal structure research. Additionally, the applications of Rpi in rare sugar production and the role of Rpi in trypanocidal drug design are reviewed. Key points • Fundamental properties of various ribose-5-phosphate isomerases (Rpis). • Differences in crystal structure and catalytic mechanism between RpiA and RpiB. • Application of Rpi as a rare sugar producer and a potential drug target.


Asunto(s)
Isomerasas Aldosa-Cetosa/química , Isomerasas Aldosa-Cetosa/metabolismo , Isomerasas Aldosa-Cetosa/clasificación , Animales , Sitios de Unión , Biocatálisis , Cristalografía por Rayos X , Humanos , Isomerismo , Cinética , Modelos Moleculares , Enfermedades Parasitarias/tratamiento farmacológico , Plantas/enzimología , Ribosamonofosfatos/metabolismo
7.
J Eukaryot Microbiol ; 63(3): 326-39, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26566594

RESUMEN

Euglenids are an ancient lineage that may have existed as early as 2 billion years ago. A mere 65 years ago, Melvin Calvin and Andrew A. Benson performed experiments on Euglena gracilis and elucidated the series of reactions by which carbon was fixed and reduced during photosynthesis. However, the evolutionary history of this pathway (Calvin-Benson cycle) in euglenids was more complex than Calvin and Benson could have imagined. The chloroplast present today in euglenophytes arose from a secondary endosymbiosis between a phagotrophic euglenid and a prasinophyte green alga. A long period of evolutionary time existed before this secondary endosymbiotic event took place, which allowed for other endosymbiotic events or gene transfers to occur prior to the establishment of the green chloroplast. This research revealed the evolutionary history of the major enzymes of the Calvin-Benson cycle throughout the euglenid lineage and showed that the majority of genes for Calvin-Benson cycle enzymes shared an ancestry with red algae and/or chromophytes suggesting they may have been transferred to the nucleus prior to the acquisition of the green chloroplast.


Asunto(s)
Evolución Biológica , Euglénidos/enzimología , Euglénidos/genética , Fotosíntesis/fisiología , Isomerasas Aldosa-Cetosa/clasificación , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/metabolismo , Teorema de Bayes , Chlorophyta/enzimología , Chlorophyta/genética , Chlorophyta/fisiología , Cloroplastos/genética , Enzimas/clasificación , Enzimas/genética , Enzimas/metabolismo , Euglénidos/metabolismo , Fructosa-Bifosfatasa/clasificación , Fructosa-Bifosfatasa/genética , Fructosa-Bifosfatasa/metabolismo , Transferencia de Gen Horizontal , Gliceraldehído-3-Fosfato Deshidrogenasas/clasificación , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Monoéster Fosfórico Hidrolasas/clasificación , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fotosíntesis/genética , Filogenia , Rhodophyta/enzimología , Simbiosis , Triosa-Fosfato Isomerasa/clasificación , Triosa-Fosfato Isomerasa/genética , Triosa-Fosfato Isomerasa/metabolismo
8.
Int J Cancer ; 137(1): 104-15, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25429733

RESUMEN

The deregulated nonoxidative pentose phosphate pathway (PPP) is known to promote oncogenesis, but the molecular mechanism remains unknown. Here, we report that human ribose-5-phosphate isomerase A (RPIA) plays a role in human hepatocellular carcinoma (HCC). A significant increase in RPIA expression was detected both in tumor biopsies of HCC patients and in a liver cancer tissue array. Importantly, the clinicopathological analysis indicated that RPIA mRNA levels were highly correlated with clinical stage, grade, tumor size, types, invasion and alpha-fetoprotein levels in the HCC patients. In addition, we demonstrated that the ability of RPIA to regulate cell proliferation and colony formation in different liver cancer cell lines required ERK signaling as well as the negative modulation of PP2A activity and that the effects of RPIA could be modulated by the addition of either a PP2A inhibitor or activator. Furthermore, the xenograft studies in nude mice revealed that the modulation of RPIA in liver cancer cells regulated tumor growth and that NIH3T3 cells overexpressing RPIA exhibited increased proliferation, enhanced colony formation, elevated levels of p-ERK1/2 and accelerated tumor growth. This study provides new insight into the molecular mechanisms by which RPIA overexpression can induce oncogenesis in HCC. Furthermore, it suggests that RPIA can be a good prognosis biomarker and a potential target for HCC therapy.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas , Isomerasas Aldosa-Cetosa/genética , Animales , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Ratones , Ratones Desnudos , Células 3T3 NIH , Trasplante de Neoplasias , Proteína Fosfatasa 2/metabolismo
9.
Food Sci Biotechnol ; 33(7): 1641-1649, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38623425

RESUMEN

Enzymatic preparation of rare sugars as an alternative to traditional sweeteners is an effective strategy to achieve a low-calorie healthy diet. Ribose-5-phosphate isomerase B (RpiB) is a key enzyme in the non-oxidative branch of the catalytic pentose phosphate pathway. Here, we investigated the potential of Curtobacterium flaccumfaciens ZXL1 (C. flaccumfaciens ZXL1) derived RpiB (CfRpiB) in D-allose preparation. The optimal reaction conditions for recombinant CfRpiB were found experimentally to be pH 7.0, 55 °C, and no metal ions. The kinetic parameters Km, kcat, and catalytic efficiency kcat/Km were 320 mM, 4769 s-1, and 14.9 mM-1 s-1 respectively. The conversion of D-allulose by purified enzyme (1 g L-1 ) to D-allose was 13% within 1 h. In addition, homology modeling and molecular docking were used to predict the active site residues: Asp13, Asp14, Cys72, Gly73, Thr74, Gly77, Asn106, and Lys144.

10.
Antioxidants (Basel) ; 12(1)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36670987

RESUMEN

Deregulation of redox homeostasis is often associated with an accelerated aging process. Ribose-5-phosphate isomerase A (RPIA) mediates redox homeostasis in the pentose phosphate pathway (PPP). Our previous study demonstrated that Rpi knockdown boosts the healthspan in Drosophila. However, whether the knockdown of rpia-1, the Rpi ortholog in Caenorhabditis elegans, can improve the healthspan in C. elegans remains unknown. Here, we report that spatially and temporally limited knockdown of rpia-1 prolongs lifespan and improves the healthspan in C. elegans, reflecting the evolutionarily conserved phenotypes observed in Drosophila. Ubiquitous and pan-neuronal knockdown of rpia-1 both enhance tolerance to oxidative stress, reduce polyglutamine aggregation, and improve the deteriorated body bending rate caused by polyglutamine aggregation. Additionally, rpia-1 knockdown temporally in the post-developmental stage and spatially in the neuron display enhanced lifespan. Specifically, rpia-1 knockdown in glutamatergic or cholinergic neurons is sufficient to increase lifespan. Importantly, the lifespan extension by rpia-1 knockdown requires the activation of autophagy and AMPK pathways and reduced TOR signaling. Moreover, the RNA-seq data support our experimental findings and reveal potential novel downstream targets. Together, our data disclose the specific spatial and temporal conditions and the molecular mechanisms for rpia-1 knockdown-mediated longevity in C. elegans. These findings may help the understanding and improvement of longevity in humans.

11.
Appl Biochem Biotechnol ; 194(10): 4852-4866, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35670905

RESUMEN

L-ribulose, a kind of high-value rare sugar, could be utilized to manufacture L-form sugars and antiviral drugs, generally produced from L-arabinose as a substrate. However, the production of L-ribulose from L-arabinose is limited by the equilibrium ratio of the catalytic reaction, hence, it is necessary to explore a new biological enzymatic method to produce L-ribulose. Ribose-5-phosphate isomerase (Rpi) is an enzyme that can catalyze the reversible isomerization between L-ribose and L-ribulose, which is of great significance for the preparation of L-ribulose. In order to obtain highly active ribose-5-phosphate isomerase to manufacture L-ribulose, ribose-5-phosphate isomerase A (OsRpiA) from Ochrobactrum sp. CSL1 was engineered based on structural and sequence analyses. Through a rational design strategy, a triple-mutant strain A10T/T32S/G101N with 160% activity was acquired. The enzymatic properties of the mutant were systematically investigated, and the optimum conditions were characterized to achieve the maximum yield of L-ribulose. Kinetic analysis clarified that the A10T/T32S/G101N mutant had a stronger affinity for the substrate and increased catalytic efficiency. Furthermore, molecular dynamics simulations indicated that the binding of the substrate to A10T/T32S/G101N was more stable than that of wild type. The shorter distance between the catalytic residues of A10T/T32S/G101N and L-ribose illuminated the increased activity. Overall, the present study provided a solid basis for demonstrating the complex functions of crucial residues in RpiAs as well as in rare sugar preparation.


Asunto(s)
Isomerasas Aldosa-Cetosa , Ochrobactrum , Isomerasas Aldosa-Cetosa/metabolismo , Antivirales , Arabinosa/metabolismo , Cinética , Ochrobactrum/genética , Ochrobactrum/metabolismo , Pentosas , Ribosa
12.
Enzyme Microb Technol ; 148: 109789, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34116752

RESUMEN

Ribose-5-phosphate isomerase A (RpiA) is of great importance in biochemistry research, however its application in biotechnology has not been fully explored. In this study the activity of RpiA from Ochrobactrum sp. CSL1 (OsRpiA) towards D-allose was engineered based on sequential and structural analyses. Strategies of alanine scanning, rational design and saturated mutagenesis were employed to create three mutant libraries. A single mutant of K124A showed a 45 % activity improvement towards D-allose. The reaction properties of the mutant were analyzed, and a shift of optimal pH and higher thermal stability at low reaction temperatures were identified. The conversion of D-allose was also improved by 40 % using K124A, and higher activities on major substrates were found in the mutant's substrate scope, implying its application potential in rare sugar preparation. Kinetics analysis revealed that Km of K124A mutant decreased by 12 % and the catalytic efficiency increased by 65 % towards D-allose. Moreover, molecular dynamics simulation illustrated the binding of substrate and K124A was more stable than that of the wild-type. The shorter distance and more relax bond angle between the catalytic residue of K124A and D-allose explained the activity improvement in detail. This study highlights the potential of OsRpiA as a biocatalyst for rare sugar preparation, and provides distinct evidences for its catalytic mechanism.


Asunto(s)
Isomerasas Aldosa-Cetosa , Ochrobactrum , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/metabolismo , Isomerismo , Ochrobactrum/metabolismo , Azúcares
13.
J Biomol Struct Dyn ; 38(7): 2047-2056, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31184542

RESUMEN

The Trypanosoma cruzi ribose-5-phosphate isomerase B (TcRpiB) is a crucial piece in the pentose phosphate pathway and thus is a potential drug target for treatment of Chagas' disease. TcRpiB residues, such as Cys69, Asp45, Glu149 and Pro47, have confirmed their roles in substrate recognition, catalytic reaction and binding site conformation. However, the joint performance of His11 and His102, in the D-ribose-5-phosphate (R5P) in the catalysis is not well understood. In this work, we probed the influence of different protonation states of His11 and His102 on the behavior of the ligand R5P using molecular dynamics simulations, network analysis and thermodynamic integration. Simulations revealed that a protonated His11 combined with a neutral His102 (His11+‒His102) was able to stabilize the ligand R5P in the binding site. Moreover, calculated relative free energy differences showed that when protonated His11 was coupled to a neutral His102 an exergonic process takes place. On the other hand, neutral His11 combined with a protonated His102 (His11‒His102+), sampled conformations that resembled the catalyzed product D-ribulose-5-phosphate (Ru5P). Network analysis also demonstrated some peculiarities for these systems with some negatively correlated nodes in the binding site for His11‒His102+, and exclusive suboptimal paths for His11+‒His102. Therefore, the combined approach presented in this paper proposes two suitable protonation states for the TcRpiB catalytic mechanism, where an extra proton in either histidines might favor R5P binding or influence isomerization reaction to Ru5P. Our results may guide further in silico drug discovery studies. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Isomerasas Aldosa-Cetosa/química , Trypanosoma cruzi , Sitios de Unión , Trypanosoma cruzi/enzimología
14.
Enzyme Microb Technol ; 140: 109604, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32912675

RESUMEN

Rare sugars have received increasing attention due to their important applications as sweeteners and building blocks. The substrate specificity and catalytic properties of ribose-5-phosphate isomerase A (RpiA) in isomerization of rare sugars have not been extensively explored. In this study, an RpiA from Ochrobactrum sp. CSL1 was cloned and expressed in Escherichia coli. The biochemical and reaction features were explored and its broad substrate specificity was identified. A higher reaction rate in isomerizing l-rhamnose to l-rhamnulose by OsRpiA, compared with OsRpiB found in the same strain indicated higher efficiency in preparing rare sugars, which was verified by kinetics study. The 2.8 Šresolution structure of OsRpiA was then solved and used in subsequent molecular dynamics experiments, providing a possible explanation for its distinct substrate specificity. The present study highlighted the unique role of microbial RpiA in preparing rare sugars, and its structural information provided a reliable reference for further reaction mechanism research and enzyme engineering work.


Asunto(s)
Isomerasas Aldosa-Cetosa/química , Isomerasas Aldosa-Cetosa/metabolismo , Ochrobactrum/enzimología , Azúcares/metabolismo , Isomerasas Aldosa-Cetosa/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Biocatálisis , Cristalografía por Rayos X , Escherichia coli/genética , Isomerismo , Cinética , Modelos Moleculares , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ramnosa/química , Ramnosa/metabolismo , Especificidad por Sustrato , Azúcares/química
15.
Sci China Life Sci ; 63(9): 1394-1405, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32157557

RESUMEN

Cancer cells remodel their metabolic network to adapt to variable nutrient availability. Pentose phosphate pathway (PPP) plays protective and biosynthetic roles by oxidizing glucose to generate reducing power and ribose. How cancer cells modulate PPP activity in response to glucose supply remains unclear. Here we show that ribose-5-phosphate isomerase A (RPIA), an enzyme in PPP, directly interacts with co-activator associated arginine methyltransferase 1 (CARM1) and is methylated at arginine 42 (R42). R42 methylation up-regulates the catalytic activity of RPIA. Furthermore, glucose deprivation strengthens the binding of CARM1 with RPIA to induce R42 hypermethylation. Insufficient glucose supply links to RPIA hypermethylation at R42, which increases oxidative PPP flux. RPIA methylation supports ROS clearance by enhancing NADPH production and fuels nucleic acid synthesis by increasing ribose supply. Importantly, RPIA methylation at R42 significantly potentiates colorectal cancer cell survival under glucose starvation. Collectively, RPIA methylation connects glucose availability to nucleotide synthesis and redox homeostasis.


Asunto(s)
Isomerasas Aldosa-Cetosa/metabolismo , Arginina/química , Neoplasias Colorrectales/metabolismo , Glucosa/metabolismo , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Inactivación de Genes , Humanos , Metilación , Ratones , Ratones Desnudos , NADP/metabolismo , Oxidación-Reducción , Vía de Pentosa Fosfato , Unión Proteica , Proteína-Arginina N-Metiltransferasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
16.
J Agric Food Chem ; 68(11): 3539-3547, 2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-32100533

RESUMEN

Ribose-5-phosphate isomerase B is of great importance for biocatalysis and biosynthesis, but the multifunctional residues in active sites hinder the research efforts. This study employed rational design strategies to locate the key residues of RpiB from Ochrobactrum sp. CSL1 (OsRpiB). A single-mutant S9T of a noncontact residue showed 80% activity improvement toward d-allose. A double-mutant S98H/S134H further increased the activity to 3.6-fold. The mutations were analyzed by kinetics and molecular dynamics analyses, indicating that S9T might enhance the substrate binding and catalysis by inducing a steric effect, and S98H/S134H could strengthen both ring opening and binding of d-allose. Though S98H/S134H showed low temperature stability, its potential was explored by isomerizing d-allose to d-psicose with higher conversion and in less reaction time. The findings of this study were beneficial for illustrating the complex functions of key residues in RpiBs and applying OsRpiB in preparing rare sugars.


Asunto(s)
Isomerasas Aldosa-Cetosa , Ochrobactrum , Isomerasas Aldosa-Cetosa/genética , Isomerasas Aldosa-Cetosa/metabolismo , Glucosa , Isomerismo , Ochrobactrum/metabolismo , Especificidad por Sustrato
17.
Eur J Med Genet ; 62(8): 103708, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31247379

RESUMEN

Ribose 5-phosphate isomerase deficiency is a rare genetic leukoencephalopathy caused by pathogenic sequence variants in RPIA, that encodes ribose 5-phosphate isomerase, an enzyme in the pentose phosphate pathway. Till date, only three individuals with ribose 5-phosphate isomerase deficiency have been described in literature. We report on a subject with RPIA associated progressive leukoencephalopathy with elevated urine arabitol and ribitol levels and a novel missense variant c.770T > C p.(Ile257Thr) in exon 8 of RPIA. We also compare the phenotypes of all the four subjects. Our report confirms the phenotype and the genetic cause of this condition.


Asunto(s)
Isomerasas Aldosa-Cetosa/deficiencia , Errores Innatos del Metabolismo de los Carbohidratos/genética , Leucoencefalopatías/genética , Polineuropatías/genética , Isomerasas Aldosa-Cetosa/genética , Alelos , Errores Innatos del Metabolismo de los Carbohidratos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Carbohidratos/patología , Humanos , Leucoencefalopatías/tratamiento farmacológico , Leucoencefalopatías/patología , Masculino , Vía de Pentosa Fosfato/genética , Polineuropatías/tratamiento farmacológico , Polineuropatías/patología , Ribitol/administración & dosificación , Alcoholes del Azúcar/administración & dosificación
18.
Cell Rep ; 28(8): 1971-1980.e8, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433975

RESUMEN

Reprogrammed metabolism and cell cycle dysregulation are two cancer hallmarks. p16 is a cell cycle inhibitor and tumor suppressor that is upregulated during oncogene-induced senescence (OIS). Loss of p16 allows for uninhibited cell cycle progression, bypass of OIS, and tumorigenesis. Whether p16 loss affects pro-tumorigenic metabolism is unclear. We report that suppression of p16 plays a central role in reprogramming metabolism by increasing nucleotide synthesis. This occurs by activation of mTORC1 signaling, which directly mediates increased translation of the mRNA encoding ribose-5-phosphate isomerase A (RPIA), a pentose phosphate pathway enzyme. p16 loss correlates with activation of the mTORC1-RPIA axis in multiple cancer types. Suppression of RPIA inhibits proliferation only in p16-low cells by inducing senescence both in vitro and in vivo. These data reveal the molecular basis whereby p16 loss modulates pro-tumorigenic metabolism through mTORC1-mediated upregulation of nucleotide synthesis and reveals a metabolic vulnerability of p16-null cancer cells.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Nucleótidos/metabolismo , Isomerasas Aldosa-Cetosa/metabolismo , Animales , Línea Celular , Senescencia Celular , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones SCID , Vía de Pentosa Fosfato , Biosíntesis de Proteínas
19.
J Child Neurol ; 33(12): 784-787, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30088433

RESUMEN

Ribose-5-phosphate isomerase deficiency, a disorder of the pentose phosphate shunt, was described in 1999. There are 2 previously reported cases of ribose-5-phosphate isomerase deficiency. Here, we describe the clinical course, diagnostic odyssey, and molecular findings in the third case of ribose-5-phosphate isomerase deficiency to further delineate the syndrome. Whole-exome sequencing demonstrated 2 mutations in the ribose-5-phosphate isomerase gene, RPIA, in a child with neonatal onset leukoencephalopathy and psychomotor delays. Urine polyols were elevated confirming deficiency of ribose-5-phosphate isomerase (RPI, EC. 5.3.1.6) and pathogenicity of the variants. Measurement of urine polyols should be considered in cases of early-onset white-matter disease.


Asunto(s)
Isomerasas Aldosa-Cetosa/deficiencia , Errores Innatos del Metabolismo de los Carbohidratos/genética , Leucoencefalopatías/genética , Mutación/genética , Polineuropatías/genética , Isomerasas Aldosa-Cetosa/genética , Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico por imagen , Preescolar , Discapacidades del Desarrollo/etiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Humanos , Leucoencefalopatías/complicaciones , Leucoencefalopatías/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Polineuropatías/complicaciones , Polineuropatías/diagnóstico por imagen , Proteínas de Transporte Vesicular
20.
J Microbiol Biotechnol ; 28(7): 1122-1132, 2018 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-29913551

RESUMEN

In this study, we attempted to find new and efficient microbial enzymes for producing rare sugars. A ribose-5-phosphate isomerase B (OsRpiB) was cloned, overexpressed, and preliminarily purified successfully from a newly screened Ochrobactrum sp. CSL1, which could catalyze the isomerization reaction of rare sugars. A study of its substrate specificity showed that the cloned isomerase (OsRpiB) could effectively catalyze the conversion of L-rhamnose to L-rhamnulose, which was unconventional for RpiB. The optimal reaction conditions (50°C, pH 8.0, and 1 mM Ca2+) were obtained to maximize the potential of OsRpiB in preparing L-rhamnulose. The catalytic properties of OsRpiB, including Km, kcat, and catalytic efficiency (kcat/Km), were determined as 43.47 mM, 129.4 sec-1, and 2.98 mM/sec. The highest conversion rate of L-rhamnose under the optimized conditions by OsRpiB could reach 26% after 4.5 h. To the best of our knowledge, this is the first successful attempt of the novel biotransformation of L-rhamnose to L-rhamnulose by OsRpiB biocatalysis.


Asunto(s)
Isomerasas Aldosa-Cetosa/química , Isomerasas Aldosa-Cetosa/metabolismo , Ochrobactrum/enzimología , Ramnosa/metabolismo , Isomerasas Aldosa-Cetosa/genética , Secuencia de Aminoácidos , Dominio Catalítico , Clonación Molecular , ADN Bacteriano/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Cinética , Ochrobactrum/aislamiento & purificación , Conformación Proteica , Proteínas Recombinantes , Ramnosa/química , Alineación de Secuencia , Análisis de Secuencia de Proteína , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda