Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Theranostics ; 11(8): 3540-3551, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664846

RESUMEN

Rationale: Aldehyde dehydrogenase (ALDH) enzymes are often upregulated in cancer cells and associated with therapeutic resistance. ALDH enzymes protect cells by metabolizing toxic aldehydes which can induce DNA double stand breaks (DSB). We recently identified a novel ALDH1A family inhibitor (ALDHi), 673A. We hypothesized that 673A, via inhibition of ALDH1A family members, could induce intracellular accumulation of genotoxic aldehydes to cause DSB and that ALDHi could synergize with inhibitors of the ATM and ATR, proteins which direct DSB repair. Methods: We used immunofluorescence to directly assess levels of the aldehyde 4-hydroxynonenal and comet assays to evaluate DSB. Western blot was used to evaluate activation of the DNA damage response pathways. Cell counts were performed in the presence of 673A and additional aldehydes or aldehyde scavengers. ALDH inhibition results were confirmed using ALDH1A3 CRISPR knockout. Synergy between 673A and ATM or ATR inhibitors was evaluated using the Chou-Talalay method and confirmed in vivo using cell line xenograft tumor studies. Results: The ALDHi 673A cellular accumulation of toxic aldehydes which induce DNA double strand breaks. This is exacerbated by addition of exogenous aldehydes such as vitamin-A (retinaldehyde) and ameliorated by aldehyde scavengers such as metformin and hydralazine. Importantly, ALDH1A3 knockout cells demonstrated increased sensitivity to ATM/ATR inhibitors. And, ALDHi synergized with inhibitors of ATM and ATR, master regulators of the DSB DNA damage response, both in vitro and in vivo. This synergy was evident in homologous recombination (HR) proficient cell lines. Conclusions: ALDHi can be used to induce DNA DSB in cancer cells and synergize with inhibitors the ATM/ATR pathway. Our data suggest a novel therapeutic approach to target HR proficient ovarian cancer cells.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Daño del ADN , Inhibidores Enzimáticos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Aldehídos/metabolismo , Aldehídos/toxicidad , Animales , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Femenino , Técnicas de Inactivación de Genes , Humanos , Ratones , Medicina de Precisión , Inhibidores de Proteínas Quinasas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biotechnol Appl Biochem ; 68(4): 744-755, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32683722

RESUMEN

Ethylene glycol and glycolic acid are bulk chemicals with a broad range of applications. The ethylene glycol and glycolic acid biosynthesis pathways have been produced by microorganisms and used as a biological route for their production. Unlike the methods that use xylose or glucose as carbon sources, xylonic acid was used as a carbon source to produce ethylene glycol and glycolic acid in this study. Amounts of 4.2 g/L of ethylene glycol and 0.7 g/L of glycolic acid were produced by a wild-type Escherichia coli W3110 within 10 H of cultivation with a substrate conversion ratio of 0.5 mol/mol. Furthermore, E. coli strains that produce solely ethylene glycol or glycolic acid were constructed. 10.3 g/L of glycolic acid was produced by E. coli ΔyqhD+aldA, and the achieved conversion ratio was 0.56 mol/mol. Similarly, the E. coli ΔaldA+yqhD produced 8.0 g/L of ethylene glycol with a conversion ratio of 0.71 mol/mol. Ethylene glycol and glycolic acid production by E. coli on xylonic acid as a carbon source provides new information on the biosynthesis pathway of these products and opens a novel way of biomass utilization.


Asunto(s)
Escherichia coli/metabolismo , Glicol de Etileno/metabolismo , Glicolatos/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/metabolismo , Aldehído Reductasa/genética , Aldehído Reductasa/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Eliminación de Gen
4.
Development ; 148(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33298461

RESUMEN

Vertebrate axial skeletal patterning is controlled by co-linear expression of Hox genes and axial level-dependent activity of HOX protein combinations. MEIS transcription factors act as co-factors of HOX proteins and profusely bind to Hox complex DNA; however, their roles in mammalian axial patterning remain unknown. Retinoic acid (RA) is known to regulate axial skeletal element identity through the transcriptional activity of its receptors; however, whether this role is related to MEIS/HOX activity remains unknown. Here, we study the role of Meis in axial skeleton formation and its relationship to the RA pathway in mice. Meis elimination in the paraxial mesoderm produces anterior homeotic transformations and rib mis-patterning associated to alterations of the hypaxial myotome. Although Raldh2 and Meis positively regulate each other, Raldh2 elimination largely recapitulates the defects associated with Meis deficiency, and Meis overexpression rescues the axial skeletal defects in Raldh2 mutants. We propose a Meis-RA-positive feedback loop, the output of which is Meis levels, that is essential to establish anterior-posterior identities and patterning of the vertebrate axial skeleton.


Asunto(s)
Tipificación del Cuerpo , Huesos/embriología , Huesos/metabolismo , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Tretinoina/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/metabolismo , Alelos , Animales , Tipificación del Cuerpo/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Feto/metabolismo , Proteínas de Homeodominio/genética , Integrasas/metabolismo , Ratones , Modelos Biológicos , Mutación/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
5.
Biochem Pharmacol ; 176: 113885, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32112881

RESUMEN

The downregulation of the denitrosylating enzyme S-nitrosoglutathione reductase (GSNOR, EC:1.1.1.284), is a feature of hepatocellular carcinoma (HCC). This condition causes mitochondrial rearrangements that sensitize these tumors to mitochondrial toxins, in particular to the mitochondrial complex II inhibitor alpha-tocopheryl succinate (αTOS). It has also been reported the GSNOR depletion impairs the selective degradation of mitochondria through mitophagy; however, if this contributes to GSNOR-deficient HCC cell sensitivity to αTOS and can be applied to anticancer therapies, is still not known. Here, we provide evidence that GSNOR-deficient HCC cells show defective mitophagy which contributes to αTOS toxicity. Mitophagy inhibition by Parkin (EC: 2.3.2.31) depletion enhances αTOS anticancer effects, thus suggesting that this drug could be effective in treating mitophagy-defective tumors.


Asunto(s)
Aldehído Oxidorreductasas/deficiencia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Mitofagia/efectos de los fármacos , alfa-Tocoferol/farmacología , Aldehído Oxidorreductasas/genética , Antioxidantes/farmacología , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Interferencia de ARN
6.
Chem Biol Interact ; 316: 108919, 2020 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-31846616

RESUMEN

Ethanol (EtOH) is a recreationally ingested compound that is both teratogenic and carcinogenic in humans. Because of its abundant consumption worldwide and the vital role of stem cells in the formation of birth defects and cancers, delineating the effects of EtOH on stem cell function is currently an active and urgent pursuit of scientific investigation to explicate some of the mechanisms contributing to EtOH toxicity. Stem cells represent a primordial, undifferentiated phase of development; thus encroachment on normal physiologic processes of differentiation into terminal lineages by EtOH can greatly alter the function of progenitors and terminally differentiated cells, leading to pathological consequences that manifest as fetal alcohol spectrum disorders and cancers. In this review we explore the disruptive role of EtOH in differentiation of stem cells. Our primary objective is to elucidate the mechanisms by which EtOH alters differentiation-related gene expression and lineage specifications, thus modifying stem cells to promote pathological outcomes. We additionally review the effects of a reactive metabolite of EtOH, acetaldehyde (AcH), in causing both differentiation defects in stem cells as well as genomic damage that incites cellular aging and carcinogenesis.


Asunto(s)
Acetaldehído/farmacología , Diferenciación Celular/efectos de los fármacos , Etanol/farmacología , Acetaldehído/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Daño del ADN/efectos de los fármacos , Etanol/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
7.
ACS Synth Biol ; 8(11): 2483-2493, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31603652

RESUMEN

Formaldehyde (HCHO) is an important intermediate in the metabolism of one-carbon (C1) compounds such as methanol, formate, and methane. The ribulose monophosphate (RuMP) pathway is the most-studied HCHO assimilation route and the 3-hexulose-6-phosphate synthase (Hps) plays an important role for HCHO fixation. In this study, we proposed and selected a pyruvate-dependent aldolase to channel HCHO into 2-keto-4-hydroxybutyrate as an important intermediate for biosynthesis. By combining this reaction with three further enzymes we demonstrated a pyruvate-based C1 metabolic pathway for biosynthesis of the appealing compound 1,3-propanediol (1,3-PDO). This novel pathway is first confirmed in vitro using HCHO and pyruvate as substrates. It is then demonstrated in vivo in E. coli for 1,3-PDO production from HCHO and methanol with glucose as a cosubstrate. This de novo pathway has several decisive advantages over the known metabolic pathways for 1,3-PDO: (1) C1 carbon is directly channeled into a precursor of 1,3-PDO; (2) the use of pyruvate as an acceptor of HCHO is glycerol-independent, circumventing thus the need of coenzyme B12 as cofactor for glycerol dehydration; (3) the pathway is much shorter and more simple than the recently proposed l-homoserine-dependent pathway, thus avoiding complicated regulations involving precursors for essential amino acids. In addition to proof-of-concept we further improved the host strain by deleting a gene (frmA) responsible for the conversion of HCHO to formate, thereby increasing the production of 1,3-PDO from 298.3 ± 11.4 mg/L to 508.3 ± 9.1 mg/L and from 3.8 mg/L to 32.7 ± 0.8 mg/L with HCHO and methanol as cosubstrate of glucose fermentation, respectively. This work is the first study demonstrating a genetically engineered E. coli that can directly use HCHO or methanol for the synthesis of 2-keto-4-hydroxybutyrate and its further conversion to 1,3-PDO.


Asunto(s)
Escherichia coli/metabolismo , Formaldehído/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Hidroxibutiratos/metabolismo , Cetonas/metabolismo , Metanol/metabolismo , Glicoles de Propileno/metabolismo , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Formaldehído/química , Fructosa-Bifosfato Aldolasa/genética , Glucosa/metabolismo , Hidroxibutiratos/química , Cetonas/química , Cinética , Redes y Vías Metabólicas , Metanol/química , Glicoles de Propileno/química , Ácido Pirúvico/metabolismo , Especificidad por Sustrato
8.
FEMS Yeast Res ; 19(2)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30476034

RESUMEN

Candida albicans is a predominant cause of fungal infections in mucosal tissues as well as life-threatening bloodstream infections in immunocompromised patients. Within the human body, C. albicans is mostly embedded in biofilms, which provides increased resistance to antifungal drugs. The glyoxalase Glx3 is an abundant proteomic component of the biofilm extracellular matrix. Here, we document phenotypic studies of a glx3Δ null mutant concerning its role in biofilm formation, filamentation, antifungal drug resistance, cell wall integrity and virulence. First, consistent with its function as glyoxalase, the glx3 null mutant showed impaired growth on media containing glycerol as the carbon source and in the presence of low concentrations of hydrogen peroxide. Importantly, the glx3Δ mutant showed decreased fitness at 37°C and formed less biofilm as compared to wild type and a reintegrant strain. At the permissive temperature of 28°C, the glx3Δ mutant showed impaired filamentation as well as increased sensitivity to Calcofluor white, Congo red, sodium dodecyl sulfate and zymolyase, indicating subtle alterations in wall architecture even though gross quantitative compositional changes were not detected. Interestingly, and consistent with its impaired filamentation, biofilm formation and growth at 37°C, the glx3Δ mutant is avirulent. Our results underline the role of Glx3 in fungal pathogenesis and the involvement of the fungal wall in this process.


Asunto(s)
Aldehído Oxidorreductasas/genética , Biopelículas/crecimiento & desarrollo , Candida albicans/fisiología , Candida albicans/efectos de la radiación , Eliminación de Gen , Respuesta al Choque Térmico , Aldehído Oxidorreductasas/deficiencia , Animales , Candida albicans/enzimología , Candida albicans/genética , Candidiasis/microbiología , Candidiasis/patología , Pared Celular/química , Modelos Animales de Enfermedad , Calor , Hifa/crecimiento & desarrollo , Ratones Endogámicos BALB C , Análisis de Supervivencia , Virulencia
9.
Autophagy ; 14(7): 1285-1287, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30029585

RESUMEN

Mitochondrial dynamics is required to adapt the manifold functions of mitochondria to cell needs and regulate their turnover by mitophagy. Actually, only if fragmented, mitochondria are engulfed by phagophores, the precursors to autophagosomes, and subsequently degraded. This process is essential to maintain a correct and healthy number of mitochondria that, otherwise, might be harmful. They, indeed, represent the main source of reactive oxygen species that - according to the mitochondrial free radical theory of aging - can cause aging when chronically overproduced. In a recent study, we demonstrated that S-nitrosylation, the reversible modification of cysteine residues by nitric oxide (NO), hyperactivates mitochondrial fragmentation by targeting DNM1L/Drp1 (dynamin 1-like) at Cys644, but inhibits mitophagy, the concomitant occurrence of these conditions driving cell senescence. We demonstrated that cell senescence, as well as mouse and human aging are characterized by an epigenetically-driven decrease in ADH5/GSNOR (alcohol dehydrogenase 5 [class III], chi polypeptide), suggesting that ADH5 may act as new longevity gene.


Asunto(s)
Envejecimiento/metabolismo , Aldehído Oxidorreductasas/metabolismo , Mitofagia , Aldehído Oxidorreductasas/deficiencia , Animales , Ratones Transgénicos , Modelos Biológicos , Nitrosación , Especies Reactivas de Oxígeno/metabolismo
10.
J Sex Med ; 15(5): 654-661, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29606625

RESUMEN

BACKGROUND: Excess reactive oxygen species and reactive nitrogen species are implicated in male infertility and impaired spermatogenesis. AIM: To investigate the effect of excess reactive nitrogen species and nitrosative stress on testicular function and the hypothalamic-pituitary-gonadal axis using the S-nitrosoglutathione reductase-null (Gsnor-/-) mouse model. METHODS: Testis size, pup number, and epididymal sperm concentration and motility of Gsnor-/- mice were compared with those of age-matched wild-type (WT) mice. Reproductive hormones testosterone (T), luteinizing hormone (LH), and follicle-stimulating hormone were compared in Gsnor-/- and WT mice. Immunofluorescence for Gsnor-/- and WT testis was performed for 3ß-hydroxysteroid dehydrogenase and luteinizing hormone receptor (LHR) and compared. Human chorionic gonadotropin and gonadotropin-releasing hormone stimulation tests were performed to assess and compare testicular and pituitary functions of Gsnor-/- and WT mice. OUTCOMES: Evaluation of fertility and reproductive hormones in Gsnor-/- vs WT mice. Response of Gsnor-/- and WT mice to human chorionic gonadotropin and gonadotropin-releasing hormone to evaluate LH and T production. RESULTS: Gsnor-/- mice had smaller litters (4.2 vs 8.0 pups per litter; P < .01), smaller testes (0.08 vs 0.09 g; P < .01), and decreased epididymal sperm concentration (69 vs 98 × 106; P < .05) and motility (39% vs 65%; P < .05) compared with WT mice. Serum T (44.8 vs 292.2 ng/dL; P < .05) and LH (0.03 vs 0.74 ng/mL; P = .04) were lower in Gsnor-/- than in WT mice despite similar follicle-stimulating hormone levels (63.98 vs 77.93 ng/mL; P = .20). Immunofluorescence of Gsnor-/- and WT testes showed similar staining of 3ß-hydroxysteroid dehydrogenase and LHR. Human chorionic gonadotropin stimulation of Gsnor-/- mice increased serum T (>1,680 vs >1,680 ng/dL) and gonadotropin-releasing hormone stimulation increased serum LH (6.3 vs 8.9 ng/mL; P = .20) similar to WT mice. CLINICAL TRANSLATION: These findings provide novel insight to a possible mechanism of secondary hypogonadism from increased reactive nitrogen species and excess nitrosative stress. STRENGTHS AND LIMITATIONS: Limitations of this study are its small samples and variability in hormone levels. CONCLUSION: Deficiency of S-nitrosoglutathione reductase results in secondary hypogonadism, suggesting that excess nitrosative stress can affect LH production from the pituitary gland. Masterson TA, Arora H, Kulandavelu S, et al. S-Nitrosoglutathione Reductase (GSNOR) Deficiency Results in Secondary Hypogonadism. J Sex Med 2018;15:654-661.


Asunto(s)
Aldehído Oxidorreductasas/deficiencia , Hipogonadismo/etiología , Hipogonadismo/patología , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Gonadotropina Coriónica/metabolismo , Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Estrés Nitrosativo/fisiología , Recuento de Espermatozoides , Testículo/patología , Testosterona/metabolismo
12.
Am J Hum Genet ; 95(5): 602-10, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439727

RESUMEN

Rhizomelic chondrodysplasia punctata (RCDP) is a group of disorders with overlapping clinical features including rhizomelia, chondrodysplasia punctata, coronal clefts, cervical dysplasia, congenital cataracts, profound postnatal growth retardation, severe intellectual disability, and seizures. Mutations in PEX7, GNPAT, and AGPS, all involved in the plasmalogen-biosynthesis pathway, have been described in individuals with RCDP. Here, we report the identification of mutations in another gene in plasmalogen biosynthesis, fatty acyl-CoA reductase 1 (FAR1), in two families affected by severe intellectual disability, early-onset epilepsy, microcephaly, congenital cataracts, growth retardation, and spasticity. Exome analyses revealed a homozygous in-frame indel mutation (c.495_507delinsT [p.Glu165_Pro169delinsAsp]) in two siblings from a consanguineous family and compound-heterozygous mutations (c.[787C>T];[1094A>G], p.[Arg263(∗)];[Asp365Gly]) in a third unrelated individual. FAR1 reduces fatty acids to their respective fatty alcohols for the plasmalogen-biosynthesis pathway. To assess the pathogenicity of the identified mutations, we transfected human embryonic kidney 293 cells with plasmids encoding FAR1 with either wild-type or mutated constructs and extracted the lipids from the cells. We screened the lipids with gas chromatography and mass spectrometry and found that all three mutations abolished the reductase activity of FAR1, given that no fatty alcohols could be detected. We also observed reduced plasmalogens in red blood cells in one individual to a range similar to that seen in individuals with RCDP, further supporting abolished FAR1 activity. We thus expand the spectrum of clinical features associated with defects in plasmalogen biosynthesis to include FAR1 deficiency as a cause of syndromic severe intellectual disability with cataracts, epilepsy, and growth retardation but without rhizomelia.


Asunto(s)
Anomalías Múltiples/genética , Aldehído Oxidorreductasas/deficiencia , Catarata/genética , Enfermedades Carenciales/genética , Epilepsia/genética , Discapacidad Intelectual/genética , Modelos Moleculares , Aldehído Oxidorreductasas/química , Aldehído Oxidorreductasas/genética , Secuencia de Bases , Cromatografía de Gases , Enfermedades Carenciales/patología , Femenino , Genotipo , Células HEK293 , Humanos , Mutación INDEL/genética , Lípidos/análisis , Imagen por Resonancia Magnética , Masculino , Espectrometría de Masas , Datos de Secuencia Molecular , Linaje , Análisis de Secuencia de ADN , Síndrome
13.
J Biol Chem ; 289(29): 20193-9, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24895131

RESUMEN

Alcohol dehydrogenase 5 (ADH5) is a conserved enzyme for alcohol and aldehyde metabolism in mammals. Despite dynamic expression throughout neurogenesis, its role in neuronal development remains unknown. Here we present the first evidence that ADH5 is a negative regulator of neuronal differentiation. Gene expression analyses identify a constant reduction of ADH5 levels throughout neuronal development. Overexpression of ADH5 reduces both development and adult neuronal differentiation of mouse neurons. This effect depends on the catalytic activity of ADH5 and involves ADH5-mediated denitrosation of histone deacetylase 2 (HDAC2). Our results indicate that ADH5 counteracts neuronal differentiation of human neural stem cells and that this effect can be reversed by pharmacological inhibition of ADH5. Based on these observations, we propose that ADH5 is a novel suppressor of neuronal differentiation and maturation. Inhibition of ADH5 may improve adult neurogenesis in a physiological or pathological setting.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Neuronas/citología , Neuronas/enzimología , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hipocampo/citología , Hipocampo/enzimología , Histona Desacetilasa 2/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/citología , Células-Madre Neurales/enzimología , Neurogénesis/genética , Neurogénesis/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
14.
Proc Natl Acad Sci U S A ; 111(2): 845-50, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24379366

RESUMEN

Lignin is one of the main factors determining recalcitrance to enzymatic processing of lignocellulosic biomass. Poplars (Populus tremula x Populus alba) down-regulated for cinnamoyl-CoA reductase (CCR), the enzyme catalyzing the first step in the monolignol-specific branch of the lignin biosynthetic pathway, were grown in field trials in Belgium and France under short-rotation coppice culture. Wood samples were classified according to the intensity of the red xylem coloration typically associated with CCR down-regulation. Saccharification assays under different pretreatment conditions (none, two alkaline, and one acid pretreatment) and simultaneous saccharification and fermentation assays showed that wood from the most affected transgenic trees had up to 161% increased ethanol yield. Fermentations of combined material from the complete set of 20-mo-old CCR-down-regulated trees, including bark and less efficiently down-regulated trees, still yielded ∼ 20% more ethanol on a weight basis. However, strong down-regulation of CCR also affected biomass yield. We conclude that CCR down-regulation may become a successful strategy to improve biomass processing if the variability in down-regulation and the yield penalty can be overcome.


Asunto(s)
Aldehído Oxidorreductasas/deficiencia , Biocombustibles , Etanol/metabolismo , Lignina/metabolismo , Populus/metabolismo , Bélgica , Biomasa , Fermentación , Francia , Plantas Modificadas Genéticamente , Populus/genética
15.
Cell Rep ; 3(5): 1503-11, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23623500

RESUMEN

The vitamin A metabolite retinoic acid (RA) provides patterning information during vertebrate embryogenesis, but the mechanism through which RA influences limb development is unclear. During patterning of the limb proximodistal axis (upper limb to digits), avian studies suggest that a proximal RA signal generated in the trunk antagonizes a distal fibroblast growth factor (FGF) signal. However, mouse and zebrafish genetic studies suggest that loss of RA suppresses forelimb initiation. Here, using genetic and pharmacological approaches, we demonstrate that limb proximodistal patterning is not RA dependent, thus indicating that RA-FGF antagonism does not occur along the proximodistal axis of the limb. Instead, our studies show that RA-FGF antagonism acts prior to limb budding along the anteroposterior axis of the trunk lateral plate mesoderm to provide a patterning cue that guides formation of the forelimb field. These findings reconcile disparate ideas regarding RA-FGF antagonism and provide insight into how endogenous RA programs the early embryo.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Esbozos de los Miembros/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Embrión de Mamíferos/fisiología , Miembro Anterior/embriología , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/crecimiento & desarrollo , Ratones , Ratones Noqueados , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , ARN Mensajero/metabolismo , Proteínas de Dominio T Box/metabolismo , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Surgery ; 152(4): 768-75; discussion 775-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23021139

RESUMEN

BACKGROUND: Homozygous null mutation of the fibroblast growth factor receptor 2IIIb (Fgfr2IIIb) gene in mice results in 42% of embryos developing duodenal atresias. Retinaldehyde dehydrogenase 2 (Raldh2, a gene critical for the generation of retinoic acid) is expressed in the mouse duodenum during the temporal window when duodenal atresias form. Raldh2 is critical for the normal development of the pancreatoduodenal region; therefore, we were interested in the effect of a Raldh2 mutation on duodenal atresia formation. To test this, we rendered Fgfr2IIIb(-/-) embryos haploinsufficient for the Raldh2 and examined these embryos for the incidence and severity of duodenal atresia. METHODS: Control embryos, Fgfr2IIIb(-/-) mutants, and Fgfr2IIIb(-/-); Raldh2(+/-) mutants were harvested at embryonic day 18.5, genotyped, and fixed overnight. Intestinal tracts were isolated. The type and severity of duodenal atresia was documented. RESULTS: A total of 97 Fgfr2IIIb(-/-) embryos were studied; 44 had duodenal atresias, and 41 of these presented as type III. In the 70 Fgfr2IIIb(-/-); Raldh2(+/-) embryos studied, a lesser incidence of duodenal atresia was seen (15 of 70; P = .0017; Fisher exact test). Atresia severity was also decreased; there were 12 embryos with type I atresias, 3 with type II atresias, and 0 with type III atresias (P < 2.81E-013; Fisher exact test). CONCLUSION: Haploinsufficiency of Raldh2 decreases the incidence and severity of duodenal atresia in the Fgfr2IIIb(-/-) model. The ability to alter defect severity through manipulation of a single gene in a specific genetic background has potentially important implications for understanding the mechanisms by which intestinal atresias arise.


Asunto(s)
Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Obstrucción Duodenal/congénito , Obstrucción Duodenal/genética , Atresia Intestinal/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Animales , Obstrucción Duodenal/embriología , Obstrucción Duodenal/metabolismo , Femenino , Haploinsuficiencia , Imagenología Tridimensional , Hibridación in Situ , Atresia Intestinal/embriología , Atresia Intestinal/metabolismo , Masculino , Ratones , Ratones Noqueados , Penetrancia , Embarazo
17.
Int Ophthalmol ; 32(5): 495-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22932746

RESUMEN

Sjögren-Larsson syndrome is an autosomal-recessive disease caused by a deficiency of the microsomal fatty aldehyde dehydrogenase enzyme. The syndrome is defined by congenital ichthyosis, spasticity, mental retardation and ocular features. We report the case of a 10-year-old boy presenting with bilateral visual impairment and photophobia. Fundus examination showed a mark of yellow-white refractile, perifoveal crystals in each eye. Optical coherence tomography (OCT) detected focal reflective structures corresponding to clinically visible intraretinal crystals and macular macrocystoids space. This case is presented to highlight the ocular findings and to evaluate the contribution of OCT in the study of the fovea anatomic changes.


Asunto(s)
Mácula Lútea/patología , Degeneración Macular/diagnóstico , Síndrome de Sjögren-Larsson/complicaciones , Tomografía de Coherencia Óptica/métodos , Aldehído Oxidorreductasas/deficiencia , Niño , Diagnóstico Diferencial , Humanos , Degeneración Macular/etiología , Masculino , Síndrome de Sjögren-Larsson/diagnóstico , Síndrome de Sjögren-Larsson/enzimología
18.
J Inherit Metab Dis ; 35(6): 955-62, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22833178

RESUMEN

This review article gives a state-of-the-art synopsis of current pathophysiological concepts in Sjögren-Larsson syndrome (SLS) mainly based upon original research data of the authors in one of the world's largest clinical SLS study cohorts. Clinical features are discussed in order of appearance, and diagnostic tests are set out to guide the clinician toward the diagnosis SLS. Furthermore, current and future treatment strategies are discussed to render a comprehensive review of the topic.


Asunto(s)
Síndrome de Sjögren-Larsson , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Bezafibrato/uso terapéutico , Encéfalo/patología , Encéfalo/fisiopatología , Carotenoides/uso terapéutico , Ojo/patología , Femenino , Terapia Genética , Humanos , Recién Nacido , Masculino , Mutación , Embarazo , Nacimiento Prematuro , Síndrome de Sjögren-Larsson/diagnóstico , Síndrome de Sjögren-Larsson/genética , Síndrome de Sjögren-Larsson/fisiopatología , Síndrome de Sjögren-Larsson/terapia , Piel/patología
19.
BMC Syst Biol ; 6: 42, 2012 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-22583864

RESUMEN

BACKGROUND: Genome-scale metabolic networks and flux models are an effective platform for linking an organism genotype to its phenotype. However, few modeling approaches offer predictive capabilities to evaluate potential metabolic engineering strategies in silico. RESULTS: A new method called "flux balance analysis with flux ratios (FBrAtio)" was developed in this research and applied to a new genome-scale model of Clostridium acetobutylicum ATCC 824 (iCAC490) that contains 707 metabolites and 794 reactions. FBrAtio was used to model wild-type metabolism and metabolically engineered strains of C. acetobutylicum where only flux ratio constraints and thermodynamic reversibility of reactions were required. The FBrAtio approach allowed solutions to be found through standard linear programming. Five flux ratio constraints were required to achieve a qualitative picture of wild-type metabolism for C. acetobutylicum for the production of: (i) acetate, (ii) lactate, (iii) butyrate, (iv) acetone, (v) butanol, (vi) ethanol, (vii) CO2 and (viii) H2. Results of this simulation study coincide with published experimental results and show the knockdown of the acetoacetyl-CoA transferase increases butanol to acetone selectivity, while the simultaneous over-expression of the aldehyde/alcohol dehydrogenase greatly increases ethanol production. CONCLUSIONS: FBrAtio is a promising new method for constraining genome-scale models using internal flux ratios. The method was effective for modeling wild-type and engineered strains of C. acetobutylicum.


Asunto(s)
Clostridium acetobutylicum/genética , Clostridium acetobutylicum/metabolismo , Biología Computacional/métodos , Genoma Bacteriano/genética , Ingeniería Metabólica/métodos , Redes y Vías Metabólicas , Modelos Biológicos , Oxidorreductasas de Alcohol/deficiencia , Oxidorreductasas de Alcohol/genética , Aldehído Oxidorreductasas/deficiencia , Aldehído Oxidorreductasas/genética , Algoritmos , Proteínas Bacterianas/genética , Clostridium acetobutylicum/enzimología , Coenzima A Transferasas/deficiencia , Coenzima A Transferasas/genética , Técnicas de Silenciamiento del Gen , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/genética , ARN sin Sentido/genética
20.
PLoS One ; 6(11): e27624, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22110697

RESUMEN

A transgenic mouse line harbouring a ß-galacdosidase reporter gene controlled by the proximal 2 kb promoter of Hoxa3 was previously generated to investigate the regulatory cues governing Hoxa3 expression in the mouse. Examination of transgenic embryos from embryonic day (E) 8.0 to E15.5 revealed regionally restricted reporter activity in the developing heart. Indeed, transgene expression specifically delineated cells from three distinct lineages: a subpopulation of the second heart field contributing to outflow tract myocardium, the cardiac neural crest cells and the pharyngeal endoderm. Manipulation of the Retinoic Acid (RA) signaling pathway showed that RA is required for correct expression of the transgene. Therefore, this transgenic line may serve as a cardiosensor line of particular interest for further analysis of outflow tract development.


Asunto(s)
Endodermo/metabolismo , Corazón/embriología , Proteínas de Homeodominio/genética , Cresta Neural/metabolismo , Faringe/embriología , Tretinoina/farmacología , Aldehído Oxidorreductasas/deficiencia , Animales , Endodermo/citología , Endodermo/efectos de los fármacos , Expresión Génica , Corazón/fisiología , Operón Lac/genética , Masculino , Ratones , Cresta Neural/citología , Cresta Neural/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transgenes/genética , Tretinoina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...