Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 326(4): C1054-C1066, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38344798

RESUMEN

To understand the role of myo-inositol oxygenase (miox) in the osmotic regulation of Nile tilapia, its expression was analyzed in various tissues. The results showed that the expression of miox gene was highest in the kidney, followed by the liver, and was significantly upregulated in the kidney and liver under 1 h hyperosmotic stress. The relative luminescence efficiency of the miox gene transcription starting site (-4,617 to +312 bp) under hyperosmotic stress was measured. Two fragments (-1,640/-1,619 and -620/-599) could induce the luminescence activity. Moreover, the -1,640/-1,619 and -620/-599 responded to hyperosmotic stress and high-glucose stimulation by base mutation, suggesting that osmotic and carbohydrate response elements may exist in this region. Finally, the salinity tolerance of Nile tilapia was significantly reduced after the knocking down of miox gene. The accumulation of myo-inositol was affected, and the expression of enzymes in glucose metabolism was significantly reduced after the miox gene was knocked down. Furthermore, hyperosmotic stress can cause oxidative stress, and MIOX may help maintain the cell redox balance under hyperosmotic stress. In summary, MIOX is essential in osmotic regulation to enhance the salinity tolerance of Nile tilapia by affecting myo-inositol accumulation, glucose metabolism, and antioxidant performance.NEW & NOTEWORTHY Myo-inositol oxygenase (MIOX) is the rate-limiting enzyme that catalyzes the first step of MI metabolism and determines MI content in aquatic animals. To understand the role of miox in the osmotic regulation of Nile tilapia, we analyzed its expression in different tissues and its function under hyperosmotic stress. This study showed that miox is essential in osmotic regulation to enhance the salinity tolerance of Nile tilapia by affecting myo-inositol accumulation, glucose metabolism, and antioxidant performance.


Asunto(s)
Cíclidos , Animales , Cíclidos/genética , Cíclidos/metabolismo , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Antioxidantes , Inositol/metabolismo , Glucosa/metabolismo
2.
Appl Environ Microbiol ; 90(6): e0014924, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38808978

RESUMEN

Glucaric acid (GA) is a value-added chemical and can be used to manufacture food additives, anticancer drugs, and polymers. The non-genetic cell-to-cell variations in GA biosynthesis are naturally inherent, indicating the presence of both high- and low-performance cells in culture. Low-performance cells can lead to nutrient waste and inefficient production. Furthermore, myo-inositol oxygenase (MIOX) is a key rate-limiting enzyme with the problem of low stability and activity in GA production. Therefore, eliminating cell-to-cell variations and increasing MIOX stability can select high-performance cells and improve GA production. In this study, an in vivo GA bioselector was constructed based on GA biosensor and tetracycline efflux pump protein TetA to continuously select GA-efficient production strains. Additionally, the upper limit of the GA biosensor was improved to 40 g/L based on ribosome-binding site optimization, achieving efficient enrichment of GA high-performance cells. A small ubiquitin-like modifier (SUMO) enhanced MIOX stability and activity. Overall, we used the GA bioselector and SUMO-MIOX fusion in fed-batch GA production and achieved a 5.52-g/L titer in Escherichia coli, which was 17-fold higher than that of the original strain.IMPORTANCEGlucaric acid is a non-toxic valuable product that was mainly synthesized by chemical methods. Due to the problems of non-selectivity, inefficiency, and environmental pollution, GA biosynthesis has attracted significant attention. The non-genetic cell-to-cell variations and MIOX stability were both critical factors for GA production. In addition, the high detection limit of the GA biosensor was a key condition for performing high-throughput screening of GA-efficient production strains. To increase GA titer, this work eliminated the cell-to-cell variations by GA bioselector constructed based on GA biosensor and TetA, and improved the stability and activity of MIOX in the GA biosynthetic pathway through fusing the SUMO to MIOX. Finally, these approaches improved the GA production by 17-fold to 5.52 g/L at 65 h. This study represents a significant step toward the industrial application of GA biosynthetic pathways in E. coli.


Asunto(s)
Escherichia coli , Ácido Glucárico , Inositol-Oxigenasa , Inositol , Escherichia coli/genética , Escherichia coli/metabolismo , Inositol/metabolismo , Inositol-Oxigenasa/metabolismo , Inositol-Oxigenasa/genética , Ácido Glucárico/metabolismo , Ingeniería Metabólica , Técnicas Biosensibles
3.
J Ind Microbiol Biotechnol ; 50(1)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-37327078

RESUMEN

Recent work in biosensors has shown promise to enable high throughput searches through large genetic libraries. However, just as physiological limitations and lack of in-depth mechanistic knowledge can prevent us from achieving high titers in microbial systems; similar roadblocks can appear in the application of biosensors. Here, we characterized a previously developed transcription-factor (ExuR) based galacturonate biosensor for its other cognate ligand, glucuronate. Though we saw an ideal response to glucuronate from the biosensor in controlled and ideal experimental circumstances, these results began to deviate from a well-behaved system when we explored the application of the sensor to different MIOX homologs. Through modifications to circuit architecture and culture conditions, we were able to decrease this variation and use these more optimal conditions to apply the biosensor for the separation of two closely related MIOX homologs. ONE-SENTENCE SUMMARY: In this work, a transcription-factor biosensor was investigated for its potential to screen a library of myo -inositol oxygenase variants while seeking to mitigate the impact the production pathway appeared to have on the biosensor.


Asunto(s)
Técnicas Biosensibles , Factores de Transcripción , Ácido Glucurónico , Factores de Transcripción/genética , Regulación de la Expresión Génica , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Glucuronatos , Técnicas Biosensibles/métodos
4.
Int J Mol Sci ; 24(4)2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36835596

RESUMEN

Inositol depletion has been associated with diabetes and related complications. Increased inositol catabolism, via myo-inositol oxygenase (MIOX), has been implicated in decreased renal function. This study demonstrates that the fruit fly Drosophila melanogaster catabolizes myo-inositol via MIOX. The levels of mRNA encoding MIOX and MIOX specific activity are increased when fruit flies are grown on a diet with inositol as the sole sugar. Inositol as the sole dietary sugar can support D. melanogaster survival, indicating that there is sufficient catabolism for basic energy requirements, allowing for adaptation to various environments. The elimination of MIOX activity, via a piggyBac WH-element inserted into the MIOX gene, results in developmental defects including pupal lethality and pharate flies without proboscises. In contrast, RNAi strains with reduced levels of mRNA encoding MIOX and reduced MIOX specific activity develop to become phenotypically wild-type-appearing adult flies. myo-Inositol levels in larval tissues are highest in the strain with this most extreme loss of myo-inositol catabolism. Larval tissues from the RNAi strains have inositol levels higher than wild-type larval tissues but lower levels than the piggyBac WH-element insertion strain. myo-Inositol supplementation of the diet further increases the myo-inositol levels in the larval tissues of all the strains, without any noticeable effects on development. Obesity and blood (hemolymph) glucose, two hallmarks of diabetes, were reduced in the RNAi strains and further reduced in the piggyBac WH-element insertion strain. Collectively, these data suggest that moderately increased myo-inositol levels do not cause developmental defects and directly correspond to reduced larval obesity and blood (hemolymph) glucose.


Asunto(s)
Drosophila melanogaster , Inositol-Oxigenasa , Animales , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Drosophila melanogaster/genética , Inositol/metabolismo , Glucosa/metabolismo , Obesidad/metabolismo , ARN Mensajero
5.
Am J Physiol Renal Physiol ; 322(3): F344-F359, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35100813

RESUMEN

Conceivably, like other forms of acute kidney injury, cadmium-induced renal injury may also be associated with oxidative stress and various forms of cell death, including necroptosis, a form of regulated necrosis-associated cell death. Myo-inositol oxygenase (MIOX), an enzyme localized in renal proximal tubules, regulates oxidative stress and programmed cell death in various forms of renal injuries. Herein, the role and potential mechanism(s) by which MIOX potentiates cadmium-induced renal tubular damage were investigated. Overexpression of MIOX exacerbated cadmium-induced cell death and proximal tubular injury in mice, whereas MIOX gene disruption attenuated cellular damage in vitro and in vivo. Furthermore, necroptosis was observed in the renal tubular compartment, and, more importantly, it was corroborated by inhibitor experiments with necrostatin-1 (Nec-1). Coadministration of Nec-1 dampened including receptor-interacting protein kinase (RIP)1/RIP3/mixed-lineage kinase domain-like signaling, which is relevant to the process of necroptosis. Interestingly, the necroptosis induced by cadmium in tubules was modulated by MIOX expression profile. Also, the increased reactive oxygen species generation and NADPH consumption were accelerated by MIOX overexpression, and they were mitigated by Nec-1 administration. These findings suggest that MIOX-potentiated redox injury and necroptosis are intricately involved in the pathogenesis of cadmium-induced nephropathy, and this may yield novel potential therapeutic targets for amelioration of cadmium-induced kidney injury.NEW & NOTEWORTHY This is a seminal article documenting the role of myo-inositol oxygenase (MIOX), a renal proximal tubule-specific enzyme, in the exacerbation of cadmium-induced acute kidney injury by perturbing redox balance and inducing necroptosis. MIOX gene disruption or administration of necrostatin-1 (a necroptosis inhibitor) diminished cadmium-induced renal damage, in both in vitro and in vivo systems, suggesting a therapeutic potential of MIOX to attenuate necroptosis and relevant signaling pathways in cadmium-induced renal injury.


Asunto(s)
Lesión Renal Aguda , Inositol-Oxigenasa , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Cadmio/metabolismo , Cadmio/toxicidad , Femenino , Humanos , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Riñón/metabolismo , Masculino , Ratones , Necroptosis , Oxidantes
6.
Mol Biol Rep ; 49(8): 8025-8035, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35294703

RESUMEN

Myo-inositol is one of the most abundant form of inositol. The myo-inositol (MI) serves as substrate to diverse biosynthesis pathways and hence it is conserved across life forms. The biosynthesis of MI is well studied in animals. Beyond biosynthesis pathway, implications of MI pathway and enzymes hold potential implications in plant physiology and crop improvement. Myo-inositol oxygenase (MIOX) enzyme catabolize MI into D-glucuronic acid (D-GlcUA). The MIOX enzyme family is well studied across few plants. More recently, the MI associated pathway's crosstalk with other important biosynthesis and stress responsive pathways in plants has drawn attention. The overall outcome from different plant species studied so far are very suggestive that MI derivatives and associated pathways could open new directions to explore stress responsive novel metabolic networks. There are evidences for upregulation of MI metabolic pathway genes, specially MIOX under different stress condition. We also found MIOX genes getting differentially expressed according to developmental and stress signals in Arabidopsis and wheat. In this review we try to highlight the missing links and put forward a tailored view over myo-inositol oxidation pathway and MIOX proteins.


Asunto(s)
Arabidopsis , Inositol-Oxigenasa , Animales , Arabidopsis/metabolismo , Vías Biosintéticas , Inositol/metabolismo , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Oxidación-Reducción
7.
BMC Genomics ; 21(1): 284, 2020 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-32252624

RESUMEN

BACKGROUND: Ascorbic acid (Vitamin C, AsA) is an antioxidant metabolite involved in plant development and environmental stimuli. AsA biosynthesis has been well studied in plants, and MIOX is a critical enzyme in plants AsA biosynthesis pathway. However, Myo-inositol oxygenase (MIOX) gene family members and their involvement in AsA biosynthesis and response to abiotic stress remain unclear. RESULTS: In this study, five tomato genes encoding MIOX proteins and possessing MIOX motifs were identified. Structural analysis and distribution mapping showed that 5 MIOX genes contain different intron/exon patterns and unevenly distributed among four chromosomes. Besides, expression analyses indicated the remarkable expression of SlMIOX genes in different plant tissues. Furthermore, transgenic lines were obtained by over-expression of the MIOX4 gene in tomato. The overexpression lines showed a significant increase in total ascorbate in leaves and red fruits compared to control. Expression analysis revealed that increased accumulation of AsA in MIOX4 overexpression lines is possible as a consequence of the multiple genes involved in AsA biosynthesis. Myo inositol (MI) feeding in leaf and fruit implied that the Myo-inositol pathway improved the AsA biosynthesis in leaves and fruits. MIOX4 overexpression lines exhibited a better light response, abiotic stress tolerance, and AsA biosynthesis capacity. CONCLUSIONS: These results showed that MIOX4 transgenic lines contribute to AsA biosynthesis, evident as better light response and improved oxidative stress tolerance. This study provides the first comprehensive analysis of the MIOX gene family and their involvement in ascorbate biosynthesis in tomato.


Asunto(s)
Ácido Ascórbico/biosíntesis , Inositol-Oxigenasa/genética , Solanum lycopersicum/genética , Secuenciación Completa del Genoma/métodos , Secuencias de Aminoácidos , Mapeo Cromosómico , Frutas/genética , Frutas/metabolismo , Regulación de la Expresión Génica de las Plantas , Inositol-Oxigenasa/química , Inositol-Oxigenasa/metabolismo , Solanum lycopersicum/metabolismo , Familia de Multigenes , Hojas de la Planta/genética , Hojas de la Planta/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Estrés Fisiológico
8.
Int J Med Microbiol ; 310(4): 151426, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32444321

RESUMEN

Recently, a new environmental Francisella strain, Francisella sp. strain W12-1067, has been identified in Germany. This strain is negative for the Francisella pathogenicity island (FPI) but exhibits a putative alternative type VI secretion system. Some known virulence factors of Francisella are present, but the pathogenic capacity of this species is not known yet. In silico genome analysis reveals the presence of a gene cluster tentatively enabling myo-inositol (MI) utilization via a putative inositol oxygenase. Labelling experiments starting from 2H-inositol demonstrate that this gene cluster is indeed involved in the metabolism of MI. We further show that, under in vitro conditions, supply of MI increases growth rates of strain W12-1067 in the absence of glucose and that the metabolism of MI is strongly reduced in a W12-1067 mutant lacking the MI gene cluster. The positive growth effect of MI in the absence of glucose is restored in this mutant strain by introducing the complete MI gene cluster. F. novicida Fx1 is also positive for the MI metabolizing gene cluster and MI again increases growth in a glucose-free medium, in contrast to F. novicida strain U112, which is shown to be a natural mutant of the MI metabolizing gene cluster. Labelling experiments of Francisella sp. strain W12-1067 in medium T containing 13C-glucose, 13C-serine or 13C-glycerol as tracers suggest a bipartite metabolism where glucose is mainly metabolized through glycolysis, but not through the Entner-Doudoroff pathway or the pentose phosphate pathway. Carbon flux from 13C-glycerol and 13C-serine is less active, and label from these tracers is transferred mostly into amino acids, lactate and fatty acids. Together, the metabolism of Francisella sp. strain W12-1067 seems to be more related to the respective one in F. novicida rather than in F. tularensis subsp. holarctica.


Asunto(s)
Carbono/metabolismo , Francisella/genética , Francisella/metabolismo , Inositol/metabolismo , Familia de Multigenes , Aminoácidos/metabolismo , Simulación por Computador , Francisella/patogenicidad , Genoma Bacteriano , Islas Genómicas , Glucosa/metabolismo , Inositol-Oxigenasa/metabolismo , Microbiología del Agua
9.
Biotechnol Lett ; 42(10): 2035-2047, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32681381

RESUMEN

The production of wheat is severely affected by abiotic stresses such as cold, drought, salinity, and high temperature. Although constitutive promoters are frequently used to regulate the expression of alien genes, these may lead to undesirable side-effects in transgenic plants. Therefore, identification and characterization of an inducible promoter that can express transgene only when exposed to stresses are of great importance in the genetic engineering of crop plants. Previous studies have indicated the abiotic stress-responsive behavior of myo-inositol oxygenase (MIOX) gene in different plants. Here, we isolated the MIOX gene promoter from wheat (TaMIOX). The in-silico analysis revealed the presence of various abiotic stress-responsive cis-elements in the promoter region. The TaMIOX promoter was fused with the UidA reporter gene and transformed into Arabidopsis thaliana. The T3 single-copy homozygous lines were analyzed for GUS activity using histochemical and fluorometric assays. Transcript expression of TaMIOX::UidA was significantly up-regulated by heat (five fold), cold (seven fold), and drought (five fold) stresses as compared to transgenic plants grown without stress-induced conditions. The CaMV35S::UidA plants showed very high GUS activity even in normal conditions. In contrast, the TaMIOX::UidA plants showed prominent GUS activity only in stress treatments (cold, heat, and drought), which suggests the inducible behavior of the TaMIOX promoter. The substrate myo-inositol feeding assay of TaMIOX::UidA plants showed lesser GUS activity as compared to plants treated in abiotic stress conditions. Results support that the TaMIOX promoter could be used as a potential candidate for conditional expression of the transgene in abiotic stress conditions.


Asunto(s)
Arabidopsis/genética , Inositol-Oxigenasa/genética , Proteínas de Plantas/genética , Regiones Promotoras Genéticas/genética , Estrés Fisiológico/genética , Arabidopsis/metabolismo , Regulación de la Expresión Génica de las Plantas/genética , Inositol-Oxigenasa/metabolismo , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/metabolismo , Triticum/enzimología , Triticum/genética
10.
Am J Physiol Renal Physiol ; 316(2): F301-F315, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30539651

RESUMEN

Besides oxidant stress, endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of various metabolic disorders affecting the kidney. These two forms of stresses are not mutually exclusive to each other and may operate by a feedback loop in worsening the cellular injury. To attest to this contention, studies were performed to assess whether in such a setting, there is worsening of tubulointerstitial injury. We employed tunicamycin as a model of ER stress and used tubular cells and mice overexpressing myo-inositol oxygenase (MIOX), an enzyme involved in glycolytic events with excessive generation of ROS. Concomitant treatment of tunicamycin and transfection of cells with MIOX-pcDNA led to a marked generation of ROS, which was reduced by MIOX-siRNA. Likewise, an accentuated expression of ER stress sensors, GRP78, XBP1, and CHOP, was observed, which was reduced with MIOX-siRNA. These sensors were markedly elevated in MIOX-TG mice compared with WT treated with tunicamycin. This was accompanied with marked deterioration of tubular morphology, along with impairment of renal functions. Interestingly, minimal damage and elevation of ER stressors was observed in MIOX-KO mice. Downstream events that were more adversely affected in MIOX-TG mice included accentuated expression of proapoptogenic proteins, proinflammatory cytokines, and extracellular matrix constituents, although expression of these molecules was unaffected in MIOX-KO mice. Also, their tunicamycin-induced accentuated expression in tubular cells was notably reduced with MIOX-siRNA. These studies suggest that the biology of MIOX-induced oxidant stress and tunicamycin-induced ER stress are interlinked, and both of the events may feed into each other to amplify the tubulointerstitial injury.


Asunto(s)
Estrés del Retículo Endoplásmico , Inositol-Oxigenasa/metabolismo , Enfermedades Renales/enzimología , Túbulos Renales Proximales/enzimología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Humanos , Inositol-Oxigenasa/genética , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales Proximales/patología , Células LLC-PK1 , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Porcinos , Tunicamicina
11.
Am J Physiol Renal Physiol ; 314(1): F107-F121, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28931523

RESUMEN

Advanced glycation end products (AGEs) play a role in pathogenesis of diabetic nephropathy (DN). Myo-inositol oxygenase (MIOX) has been implicated in tubulointerstitial injury in the context of DN. We investigated the effect of AGEs on MIOX expression and delineated mechanisms that lead to tubulointerstitial injury. The status of MIOX, RAGE, and relevant cellular signaling pathways activated following AGE:RAGE interaction was examined in tubular cells and kidneys of AGE-BSA-treated mice. A solid-phase assay revealed an enhanced binding of RAGE with AGE-BSA, AGE-laminin, and AGE-collagen IV. The cells treated with AGE-BSA had increased MIOX activity/expression and promoter activity. This was associated with activation of various signaling kinases of phosphatidylinositol 3-kinase (PI3K)-AKT pathway and increased expression of NF-κB, transforming growth factor (TGF)-ß, and fibronectin, which was negated with the treatment of MIOX/RAGE- small interfering (si) RNA. Concomitant with MIOX upregulation, there was an increased generation of reactive oxygen species (ROS), which could be abrogated with MIOX/RAGE- siRNA treatment. The kidneys of mice treated with AGE-BSA had significantly high urinary A/C ratio, upregulation of MIOX, RAGE and NF-κB, along with influx of monocytes into the tubulointerstitium, increased the expression of MCP-1, IL-6, and fibronectin and increased the generation of ROS. Such perturbations were abrogated with the concomitant treatment of inhibitors MIOX or RAGE (d-glucarate and FPS-ZM1). These studies support a role of AGE:RAGE interaction in the activation of PI3K-AKT pathway and upregulation of MIOX, with excessive generation of ROS, increased expression of NF-κB, inflammatory cytokines, TGF-ß, and fibronectin. Collectively, these observations highlight the relevance of the biology of MIOX in the contribution toward tubulointerstitial injury in DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Fibronectinas/metabolismo , Inositol-Oxigenasa/metabolismo , Riñón/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Albúmina Sérica Bovina/metabolismo
12.
Metab Eng ; 49: 212-219, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30125674

RESUMEN

The development of D-glucaric acid (GA) production in recombinant cells has leapt forward in recent years, and higher throughput screening and selection of better-performing recombinant cells or biocatalysts is in current demand. A biosensor system which converts GA concentration into fluorescence signal in Escherichia coli was developed in 2016, but its application has rarely been reported. Herein, an effective high-throughput screening approach independent of special-purpose devices such as microfluidic platforms was established and tentatively applied. In this one-pot two-strain system, GA producers-bacterial or yeast cells containing the GA biosynthetic pathway-were sorted with the help of another E. coli strain acting as a GA biosensor. The identification of highly active mutants of myo-inositol oxygenase through this system validates its effectiveness in sorting E. coli cells. Subsequently, accurate ranking of the GA synthesis capacity of a small library of Saccharomyces cerevisiae strains containing distinct GA synthesis pathways demonstrated that this optimized one-pot two-strain system may also be used for eukaryotic producer strains. These results will assist in research into metabolic engineering for GA production and development of biosensor applications.


Asunto(s)
Técnicas Biosensibles , Escherichia coli , Glutaratos , Inositol-Oxigenasa , Mutación , Saccharomyces cerevisiae , Escherichia coli/genética , Escherichia coli/metabolismo , Glutaratos/análisis , Glutaratos/metabolismo , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
13.
Am J Pathol ; 187(4): 724-739, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28208054

RESUMEN

The catabolic enzyme myo-inositol oxygenase (MIOX) is expressed in proximal tubules and up-regulated in the diabetic state. Previously, we reported its transcriptional and translation regulation by high glucose (HG), osmolytes, and fatty acids. However, its epigenetic regulation is unknown. Bisulfite sequencing revealed that both human and mouse MIOX promoters, enriched with CpG sites, are hypomethylated and unmethylated under HG ambience and hyperglycemic states associated with increased MIOX expression. Eletrophoretic mobility shift assays revealed increased binding of unmethylated oligos with nucleoproteins of cells maintained under HG. In addition, a strong binding of specificity protein (Sp)-1 transcription factor with MIOX promoter was observed under HG, especially with unmethylated Sp-1 oligo. Specificity of binding was established by supershift assays and treatment with the Sp-1 inhibitor mithramycin. Promoter analysis revealed an increase in luciferase activity under HG, which was reduced after mutation of the Sp-1-binding site. Sp1 siRNA treatment reduced mRNA and protein expression of Sp-1 and MIOX and generation of reactive oxygen species derived from NADPH oxidase (NOX)-4 and mitochondrial sources. In addition, there was reduced expression of hypoxia-inducible factor-1α relevant in the pathogenesis of diabetic nephropathy. Sp1 siRNA treatment reduced fibronectin expression, an extracellular matrix protein that is increased in diabetic nephropathy and tubulopathy. HG-induced MIOX expression was also reduced with the treatment of apelin-13, which deacetylates histones. Overall, these findings highlight the epigenetic regulation of MIOX in the pathogenesis of diabetic tubulopathy.


Asunto(s)
Metilación de ADN/genética , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Glucosa/toxicidad , Inositol-Oxigenasa/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Islas de CpG/genética , Nefropatías Diabéticas/genética , Fibronectinas/metabolismo , Eliminación de Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inositol-Oxigenasa/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nucleoproteínas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo
14.
J Biol Chem ; 291(11): 5688-5707, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26792859

RESUMEN

Diabetic nephropathy (DN) is characterized by perturbations in metabolic/cellular signaling pathways with generation of reactive oxygen species (ROS). The ROS are regarded as a common denominator of various pathways, and they inflict injury on renal glomerular cells. Recent studies indicate that tubular pathobiology also plays a role in the progression of DN. However, the mechanism(s) for how high (25 mm) glucose (HG) ambience induces tubular damage remains enigmatic. myo-Inositol oxygenase (MIOX) is a tubular enzyme that catabolizes myo-inositol to d-glucuronate via the glucuronate-xylulose (G-X) pathway. In this study, we demonstrated that G-X pathway enzymes are expressed in the kidney, and MIOX expression/bioactivity was up-regulated under HG ambience in LLC-PK1 cells, a tubular cell line. We further investigated whether MIOX overexpression leads to accentuation of tubulo-interstitial injury, as gauged by some of the parameters relevant to the progression of DN. Under HG ambience, MIOX overexpression accentuated redox imbalance, perturbed NAD(+)/NADH ratios, increased ROS generation, depleted reduced glutathione, reduced GSH/GSSG ratio, and enhanced adaptive changes in the profile of the antioxidant defense system. These changes were also accompanied by mitochondrial dysfunctions, DNA damage and induction of apoptosis, accentuated activity of profibrogenic cytokine, and expression of fibronectin, the latter two being the major hallmarks of DN. These perturbations were largely blocked by various ROS inhibitors (Mito Q, diphenyleneiodonium chloride, and N-acetylcysteine) and MIOX/NOX4 siRNA. In conclusion, this study highlights a novel mechanism where MIOX under HG ambience exacerbates renal injury during the progression of diabetic nephropathy following the generation of excessive ROS via an unexplored G-X pathway.


Asunto(s)
Nefropatías Diabéticas/patología , Glucosa/metabolismo , Inositol-Oxigenasa/metabolismo , Riñón/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Peróxido de Hidrógeno/metabolismo , Inositol-Oxigenasa/análisis , Inositol-Oxigenasa/genética , Riñón/metabolismo , Células LLC-PK1 , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , NAD/metabolismo , Estrés Oxidativo , Porcinos , Regulación hacia Arriba
15.
J Biol Chem ; 291(3): 1348-67, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26578517

RESUMEN

The kidney is one of the target organs for various metabolic diseases, including diabetes, metabolic syndrome, and obesity. Most of the metabolic studies underscore glomerular pathobiology, although the tubulo-interstitial compartment has been underemphasized. This study highlights mechanisms concerning the pathobiology of tubular injury in the context of myo-inositol oxygenase (Miox), a tubular enzyme. The kidneys of mice fed a high fat diet (HFD) had increased Miox expression and activity, and the latter was related to phosphorylation of serine/threonine residues. Also, expression of sterol regulatory element-binding protein1 (Srebp1) and markers of cellular/nuclear damage was increased along with accentuated apoptosis and loss of tubular brush border. Similar results were observed in cells treated with palmitate/BSA. Multiple sterol-response elements and E-box motifs were found in the miox promoter, and its activity was modulated by palmitate/BSA. Electrophoretic mobility and ChIP assays confirmed binding of Srebp to consensus sequences of the miox promoter. Exposure of palmitate/BSA-treated cells to rapamycin normalized Miox expression and prevented Srebp1 nuclear translocation. In addition, rapamycin treatment reduced p53 expression and apoptosis. Like rapamycin, srebp siRNA reduced Miox expression. Increased expression of Miox was associated with the generation of reactive oxygen species (ROS) in kidney tubules of mice fed an HFD and cell exposed to palmitate/BSA. Both miox and srebp1 siRNAs reduced generation of ROS. Collectively, these findings suggest that HFD or fatty acids modulate transcriptional, translational, and post-translational regulation of Miox expression/activity and underscore Miox being a novel target of the transcription factor Srebp1. Conceivably, activation of the mTORC1/Srebp1/Miox pathway leads to the generation of ROS culminating into tubulo-interstitial injury in states of obesity.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Inositol-Oxigenasa/metabolismo , Túbulos Renales/enzimología , Obesidad/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Regulación hacia Arriba , Animales , Apoptosis , Línea Celular , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Dieta Alta en Grasa/efectos adversos , Humanos , Inositol-Oxigenasa/antagonistas & inhibidores , Inositol-Oxigenasa/genética , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Ratones , Obesidad/etiología , Obesidad/patología , Oxigenasas/antagonistas & inhibidores , Oxigenasas/genética , Oxigenasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas/antagonistas & inhibidores , Proteínas/genética , Proteínas/metabolismo , Interferencia de ARN , Ratas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Sus scrofa
16.
Proc Natl Acad Sci U S A ; 110(47): 18874-9, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24198335

RESUMEN

The founding members of the HD-domain protein superfamily are phosphohydrolases, and newly discovered members are generally annotated as such. However, myo-inositol oxygenase (MIOX) exemplifies a second, very different function that has evolved within the common scaffold of this superfamily. A recently discovered HD protein, PhnZ, catalyzes conversion of 2-amino-1-hydroxyethylphosphonate to glycine and phosphate, culminating a bacterial pathway for the utilization of environmentally abundant 2-aminoethylphosphonate. Using Mössbauer and EPR spectroscopies, X-ray crystallography, and activity measurements, we show here that, like MIOX, PhnZ employs a mixed-valent Fe(II)/Fe(III) cofactor for the O2-dependent oxidative cleavage of its substrate. Phylogenetic analysis suggests that many more HD proteins may catalyze yet-unknown oxygenation reactions using this hitherto exceptional Fe(II)/Fe(III) cofactor. The results demonstrate that the catalytic repertoire of the HD superfamily extends well beyond phosphohydrolysis and suggest that the mechanism used by MIOX and PhnZ may be a common strategy for oxidative C-X bond cleavage.


Asunto(s)
Bacterias/enzimología , Inositol-Oxigenasa/química , Inositol-Oxigenasa/metabolismo , Modelos Moleculares , Organofosfonatos/metabolismo , Conformación Proteica , Catálisis , Cristalografía por Rayos X , Escherichia coli , Inositol-Oxigenasa/genética , Estructura Molecular , Filogenia , Espectroscopía de Mossbauer
17.
J Am Soc Nephrol ; 26(6): 1304-21, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25270067

RESUMEN

Diabetic kidney disease (DKD) is associated with oxidative stress and mitochondrial injury. Myo-inositol oxygenase (MIOX), a tubular-specific enzyme, modulates redox imbalance and apoptosis in tubular cells in diabetes, but these mechanisms remain unclear. We investigated the role of MIOX in perturbation of mitochondrial quality control, including mitochondrial dynamics and autophagy/mitophagy, under high-glucose (HG) ambience or a diabetic state. HK-2 or LLC-PK1 cells subjected to HG exhibited an upregulation of MIOX accompanied by mitochondrial fragmentation and depolarization, inhibition of autophagy/mitophagy, and altered expression of mitochondrial dynamic and mitophagic proteins. Furthermore, dysfunctional mitochondria accumulated in the cytoplasm, which coincided with increased reactive oxygen species generation, Bax activation, cytochrome C release, and apoptosis. Overexpression of MIOX in LLC-PK1 cells enhanced the effects of HG, whereas MIOX siRNA or d-glucarate, an inhibitor of MIOX, partially reversed these perturbations. Moreover, decreasing the expression of MIOX under HG ambience increased PTEN-induced putative kinase 1 expression and the dependent mitofusin-2-Parkin interaction. In tubules of diabetic mice, increased MIOX expression and mitochondrial fragmentation and defective autophagy were observed. Dietary supplementation of d-glucarate in diabetic mice decreased MIOX expression, attenuated tubular damage, and improved renal functions. Notably, d-glucarate administration also partially attenuated mitochondrial fragmentation, oxidative stress, and apoptosis and restored autophagy/mitophagy in the tubular cells of these mice. These results suggest a novel mechanism linking MIOX to impaired mitochondrial quality control during tubular injury in the pathogenesis of DKD and suggest d-glucarate as a potential therapeutic agent for the amelioration of DKD.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/genética , Glucuronatos/farmacología , Inositol-Oxigenasa/genética , Túbulos Renales/metabolismo , Mitocondrias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Inmunohistoquímica , Inositol-Oxigenasa/metabolismo , Pruebas de Función Renal , Túbulos Renales/enzimología , Células LLC-PK1/efectos de los fármacos , Células LLC-PK1/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Sensibilidad y Especificidad , Estreptozocina/farmacología , Porcinos , Regulación hacia Arriba
18.
Plant Mol Biol ; 88(6): 545-60, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26129988

RESUMEN

Ascorbic acid (AsA) biosynthesis and its implications for stress tolerance and plant development were investigated in a set of rice knock-out (KO) mutants for AsA biosynthetic genes and their wild-types. KO of two isoforms of GDP-D-mannose epimerase (OsGME) reduced the foliar AsA level by 20-30%, and KO of GDP-L-galactose phosphorylase (OsGGP) by 80%, while KO of myo-inositol oxygenase (OsMIOX) did not affect foliar AsA levels. AsA concentration was negatively correlated with lipid peroxidation in foliar tissue under ozone stress and zinc deficiency, but did not affect the sensitivity to iron toxicity. Lack of AsA reduced the photosynthetic efficiency as represented by the maximum carboxylation rate of Rubisco (Vmax), the maximum electron transport rate (Jmax) and the chlorophyll fluorescence parameter ΦPSII. Mutants showed lower biomass production than their wild-types, especially when OsGGP was lacking (around 80% reductions). All plants except for KO mutants of OsGGP showed distinct peaks in foliar AsA concentrations during the growth, which were consistent with up-regulation of OsGGP, suggesting that OsGGP plays a pivotal role in regulating foliar AsA levels during different growth stages. In conclusion, our data demonstrate multiple roles of AsA in stress tolerance and development of rice.


Asunto(s)
Ácido Ascórbico/biosíntesis , Oryza/crecimiento & desarrollo , Oryza/metabolismo , Desarrollo de la Planta/fisiología , Estrés Fisiológico/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación de la Expresión Génica de las Plantas/fisiología , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Hierro , Ozono , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Zinc
19.
Am J Physiol Renal Physiol ; 309(9): F755-63, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26311112

RESUMEN

Renal depletion of myo-inositol (MI) is associated with the pathogenesis of diabetic nephropathy in animal models, but the underlying mechanisms involved are unclear. We hypothesized that MI depletion was due to changes in inositol metabolism and therefore examined the expression of genes regulating de novo biosynthesis, reabsorption, and catabolism of MI. We also extended the analyses from diabetes mellitus to animal models of dietary-induced obesity and hypertension. We found that renal MI depletion was pervasive across these three distinct disease states in the relative order: hypertension (-51%)>diabetes mellitus (-35%)>dietary-induced obesity (-19%). In 4-wk diabetic kidneys and in kidneys derived from insulin-resistant and hypertensive rats, MI depletion was correlated with activity of the MI-degrading enzyme myo-inositol oxygenase (MIOX). By contrast, there was decreased MIOX expression in 8-wk diabetic kidneys. Immunohistochemistry localized the MI-degrading pathway comprising MIOX and the glucuronate-xylulose (GX) pathway to the proximal tubules within the renal cortex. These findings indicate that MI depletion could reflect increased catabolism through MIOX and the GX pathway and implicate a common pathological mechanism contributing to renal oxidative stress in metabolic disease.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Hipertensión/metabolismo , Inositol/metabolismo , Túbulos Renales Proximales/metabolismo , Obesidad/metabolismo , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Hipertensión/complicaciones , Hipertensión/genética , Inositol/deficiencia , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Resistencia a la Insulina , Túbulos Renales Proximales/enzimología , Masculino , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/genética , Proteínas/genética , Proteínas/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Wistar , Xilulosa/genética , Xilulosa/metabolismo
20.
Metab Eng ; 22: 22-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333274

RESUMEN

D-glucaric acid has been explored for a myriad of potential uses, including biopolymer production and cancer treatment. A biosynthetic route to produce D-glucaric acid from glucose has been constructed in Escherichia coli (Moon et al., 2009b), and analysis of the pathway revealed myo-inositol oxygenase (MIOX) to be the least active enzyme. To increase pathway productivity, we explored protein fusion tags for increased MIOX solubility and directed evolution for increased MIOX activity. An N-terminal SUMO fusion to MIOX resulted in a 75% increase in D-glucaric acid production from myo-inositol. While our directed evolution efforts did not yield an improved MIOX variant, our screen isolated a 941 bp DNA fragment whose expression led to increased myo-inositol transport and a 65% increase in D-glucaric acid production from myo-inositol. Overall, we report the production of up to 4.85 g/L of D-glucaric acid from 10.8 g/L myo-inositol in recombinant E. coli.


Asunto(s)
Estabilidad de Enzimas , Escherichia coli , Ácido Glucárico/metabolismo , Inositol/metabolismo , Transporte Biológico Activo/genética , Escherichia coli/enzimología , Escherichia coli/genética , Inositol-Oxigenasa/genética , Inositol-Oxigenasa/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA