Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1882-1894, 2024 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-38914498

RESUMEN

1,4-cyclohexanedimethylamine (1,4-BAC) is an important monomer for bio-based materials, it finds wide applications in various fields including organic synthesis, medicine, chemical industry, and materials. At present, its synthesis primarily relies on chemical method, which suffer from issues such as expensive metal catalyst, harsh reaction conditions, and safety risks. Therefore, it is necessary to explore greener alternatives for its synthesis. In this study, a two-bacterium three-enzyme cascade conversion pathway was successfully developed to convert 1,4-cyclohexanedicarboxaldehyde to 1,4-cyclohexanedimethylamine. This pathway used Escherichia coli derived aminotransferase (EcTA), Saccharomyces cerevisiae derived glutamate dehydrogenase (ScGlu-DH), and Candida boidinii derived formate dehydrogenase (CbFDH). Through structure-guided protein engineering, a beneficial mutant, EcTAF91Y, was obtained, exhibiting a 2.2-fold increase in specific activity and a 1.9-fold increase in kcat/Km compared to that of the wild type. By constructing recombinant strains and optimizing reaction conditions, it was found that under the optimal conditions, a substrate concentration of 40 g/L could produce (27.4±0.9) g/L of the product, corresponding to a molar conversion rate of 67.5%±2.1%.


Asunto(s)
Escherichia coli , Saccharomyces cerevisiae , Escherichia coli/metabolismo , Escherichia coli/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/enzimología , Transaminasas/metabolismo , Transaminasas/genética , Ingeniería de Proteínas , Glutamato Deshidrogenasa/metabolismo , Glutamato Deshidrogenasa/genética , Formiato Deshidrogenasas/metabolismo , Formiato Deshidrogenasas/genética , Candida/enzimología , Candida/metabolismo , Ciclohexilaminas/metabolismo
2.
Trends Pharmacol Sci ; 44(12): 902-916, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37770317

RESUMEN

Ferroptosis is an iron-catalysed form of regulated cell death, which is critically dependent on phospholipid peroxidation of cellular membranes. Ferrostatin 1 was one of the first synthetic radical-trapping antioxidants (RTAs) reported to block ferroptosis and it is widely used as reference compound. Ferroptosis has been linked to multiple diseases and the use of its inhibitors could have therapeutic potential. Although, novel biochemical pathways provide insights for different pharmacological targets, the use of lipophilic RTAs to block ferroptosis remains superior. In this Review, we provide a comprehensive overview of the different classes of ferroptosis inhibitors, focusing on endogenous and synthetic RTAs. A thorough analysis of their chemical, pharmacokinetic, and pharmacological properties and potential for in vivo use is provided.


Asunto(s)
Ferroptosis , Humanos , Peroxidación de Lípido , Ciclohexilaminas/metabolismo , Ciclohexilaminas/farmacología , Antioxidantes/farmacología
3.
Protein Expr Purif ; 198: 106136, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35760252

RESUMEN

Paenarthrobacter sp. TYUT067 is a soil bacterium that can degrade and use cyclohexylamine as the sole source of carbon and energy. However, the responsible enzymes involved in cyclohexylamine degradation by TYUT067 have not been cloned and characterized in detail yet. In this study, four possible cyclohexylamine degradation genes, one cyclohexylamine oxidase (Pachao), two cyclohexanone monooxygenases (Pachms) and one lactone hydrolase (Pamlh) were successfully cloned and heterologous expressed in Escherichia coli T7 host cells. The four enzymes were purified and characterized. The optimal pH and temperature of the purified enzymes toward their own substrates were 7.0 (PaCHAO), 8.0 (PaCHM1), 9.0 (PaCHM2 and PaMLH) and 30 °C (PaCHAO and PaMLH), 40 °C (PaCHM2) and 45 °C (PaCHM1), respectively, with KM of 1.1 mM (PaCHAO), 0.1 mM (PaCHM1), 0.1 mM (PaCHM2) and 0.8 mM (PaMLH), and yielding a catalytic efficiency kcat/KM of 16.1 mM-1 s-1 (PaCHAO), 1.0 mM-1 s-1 (PaCHM1), 5.0 mM-1 s-1 (PaCHM2) and 124.4 mM-1 s-1 (PaMLH). In vitro mimicking the cyclohexylamine degradation pathway was conducted by using the combined three cyclohexylamine degradation enzymes (PaCHAO, PaCHM2 and PaMLH) with 10-50 mM cyclohexylamine, 100% conversion of cyclohexylamine could be finished within 12 h without any detected intermediates. The current study confirmed the enzymes responsible for cyclohexylamine degradation in TYUT067 for the first time, provide basic information for further investigation and application of these specific enzymes in pollution control.


Asunto(s)
Ciclohexilaminas , Micrococcaceae , Clonación Molecular , Ciclohexilaminas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Hidrolasas/genética , Micrococcaceae/metabolismo
4.
FEBS J ; 289(3): 671-681, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34544203

RESUMEN

Iron homeostasis is essential for both sides of the host-pathogen interface. Restricting access of iron slows bacterial growth while iron is also a necessary cofactor for host immunity. Haem oxygenase 1 (HMOX1) is a critical regulator of iron homeostasis that catalyses the liberation of iron during degradation of haem. It is also a stress-responsive protein that can be rapidly upregulated and confers protection to the host. Although a protective role of HMOX1 has been demonstrated in a variety of diseases, the role of HMOX1 in Mycobacterium tuberculosis infection is equivocal across experiments with different host-pathogen combinations. Here, we use the natural host-pathogen pairing of the zebrafish-Mycobacterium marinum infection platform to study the role of zebrafish haem oxygenase in mycobacterial infection. We identify zebrafish Hmox1a as the relevant functional paralog of mammalian HMOX1 and demonstrate a conserved role for Hmox1a in protecting the host from M. marinum infection. Using genetic and chemical tools, we show zebrafish Hmox1a protects the host against M. marinum infection by reducing infection-induced iron accumulation and ferrostatin-sensitive cell death.


Asunto(s)
Hemo-Oxigenasa 1/genética , Hierro/metabolismo , Tuberculosis/genética , Proteínas de Pez Cebra/genética , Animales , Muerte Celular/genética , Ciclohexilaminas/metabolismo , Modelos Animales de Enfermedad , Hemo/genética , Homeostasis , Interacciones Huésped-Patógeno/genética , Humanos , Macrófagos/microbiología , Infecciones por Mycobacterium no Tuberculosas , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Fenilendiaminas/metabolismo , Tuberculosis/microbiología , Pez Cebra/genética , Pez Cebra/microbiología
5.
Prostaglandins Other Lipid Mediat ; 158: 106604, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34922004

RESUMEN

Adipose tissue contains a complex immune environment and is a central contributor to heightened systemic inflammation in obese persons. Epoxyeicosatrienoic acids (EETs) are lipid signaling molecules that decrease inflammation in obese animals, but their effect on inflammation in humans is unknown. The enzyme soluble epoxide hydrolase (sEH) hydrolyzes EETs to less active diols, and we hypothesized that pharmacologic sEH inhibition would decrease adipose inflammation in obese individuals. We treated obese prediabetic adults with the sEH inhibitor GSK2256294 versus placebo in a crossover design, collected subcutaneous abdominal adipose tissue via lipoaspiration and characterized the tissue T cell profile. Treatment with GSK2256294 decreased the percentage of pro-inflammatory T cells producing interferon-gamma (IFNγ), but not interleukin (IL)-17A, and decreased the amount of secreted tumor necrosis factor-alpha (TNFα). Understanding the contribution of the EET/sEH pathway to inflammation in obesity could lead to new strategies to modulate adipose and systemic inflammation.


Asunto(s)
Epóxido Hidrolasas , Linfocitos T , Tejido Adiposo/metabolismo , Animales , Ciclohexilaminas/metabolismo , Epóxido Hidrolasas/metabolismo , Linfocitos T/metabolismo , Triazinas
6.
Toxicol In Vitro ; 73: 105146, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33737050

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and striatum. Aging is the most important risk factor of PD. Ferroptosis is an iron-dependent form of cell death associated with PD. However, it is not clear whether ferroptosis accelerates PD by promoting cellular senescence. This study investigated the mechanism of 1-methyl-4-phenylpyridinium (MPP+) -induced PC12 cells injury. We found that MPP+ induced cell senescence with increased ß-galactosidase activity and the expression of p53, p21 and p16 activation in cells. In addition, MPP+ treatment showed smaller mitochondria and increased membrane density, downregulation of ferritin heavy chain 1 expression and upregulation of acyl-CoA synthetase long chain family member 4 expression, and enhanced levels of oxidative stress, which were important characteristics of ferroptosis. Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, was tested to eliminate MPP+-induced cell senescence. Fer-1 downregulated the expression of p53 and upregulated the expression of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase-4 (GPX4) in MPP+-induced ferroptosis. Inhibition of p53 eliminated cell senescence by upregulation the expression of of SLC7A11 and GPX4. Thus, these results suggest that MPP+ induces senescence in PC12 cells via the p53/ SLC7A11/ GPX4 signaling pathway in the ferroptosis regulation mechanism.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Senescencia Celular/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Ciclohexilaminas/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Fenilendiaminas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , beta-Galactosidasa/metabolismo
7.
J Am Chem Soc ; 142(10): 4892-4903, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32114761

RESUMEN

Human ornithine aminotransferase (hOAT), a pyridoxal 5'-phosphate-dependent enzyme, plays a critical role in the progression of hepatocellular carcinoma (HCC). Pharmacological selective inhibition of hOAT has been shown to be a potential therapeutic approach for HCC. Inspired by the discovery of the nonselective aminotransferase inactivator (1R,3S,4S)-3-amino-4-fluoro cyclopentane-1-carboxylic acid (1), in this work, we rationally designed, synthesized, and evaluated a novel series of fluorine-substituted cyclohexene analogues, thereby identifying 8 and 9 as novel selective hOAT time-dependent inhibitors. Intact protein mass spectrometry and protein crystallography demonstrated 8 and 9 as covalent inhibitors of hOAT, which exhibit two distinct inactivation mechanisms resulting from the difference of a single fluorine atom. Interestingly, they share a similar turnover mechanism, according to the mass spectrometry-based analysis of metabolites and fluoride ion release experiments. Molecular dynamics (MD) simulations and electrostatic potential (ESP) charge calculations were conducted, which elucidated the significant influence of the one-fluorine difference on the corresponding intermediates, leading to two totally different inactivation pathways. The novel addition-aromatization inactivation mechanism for 9 contributes to its significantly enhanced potency, along with excellent selectivity over other aminotransferases.


Asunto(s)
Ácidos Ciclohexanocarboxílicos/química , Ciclohexilaminas/química , Inhibidores Enzimáticos/química , Hidrocarburos Fluorados/química , Ornitina-Oxo-Ácido Transaminasa/antagonistas & inhibidores , Ácidos Ciclohexanocarboxílicos/síntesis química , Ácidos Ciclohexanocarboxílicos/metabolismo , Ciclohexilaminas/síntesis química , Ciclohexilaminas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Humanos , Hidrocarburos Fluorados/síntesis química , Hidrocarburos Fluorados/metabolismo , Modelos Químicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Ornitina-Oxo-Ácido Transaminasa/química , Ornitina-Oxo-Ácido Transaminasa/metabolismo , Unión Proteica , Fosfato de Piridoxal/química , Ácido gamma-Aminobutírico/análogos & derivados
8.
Int J Cancer ; 146(2): 510-520, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31173656

RESUMEN

Recent data suggest that rhabdomyosarcoma (RMS) cells might be vulnerable to oxidative stress-induced cell death. Here, we show that RMS are susceptible to cell death induced by Erastin, an inhibitor of the glutamate/cystine antiporter xc- that can increase reactive oxygen species (ROS) production via glutathione (GSH) depletion. Prior to cell death, Erastin caused GSH depletion, ROS production and lipid peroxidation. Importantly, pharmacological inhibitors of lipid peroxidation (i.e., Ferrostatin-1, Liproxstatin-1), ROS scavengers (i.e., α-Tocopherol, GSH) and the iron chelator Deferoxamine inhibited ROS accumulation, lipid peroxidation and cell death, consistent with ferroptosis. Interestingly, the broad-spectrum protein kinase C (PKC) inhibitor Bisindolylmaleimide I as well as the PKCα- and ß-selective inhibitor Gö6976 significantly reduced Erastin-induced cell death. Similarly, genetic knockdown of PKCα significantly protected RMS cells from Erastin-induced cell death. Furthermore, the broad-spectrum nicotinamide adenine dinucleotide phosphate-oxidase (NOX) inhibitor Diphenyleneiodonium and the selective NOX1/4 isoform inhibitor GKT137831 significantly decreased Erastin-stimulated ROS, lipid ROS and cell death. These data provide new insights into the molecular mechanisms of ferroptosis in RMS, contributing to the development of new redox-based treatment strategies.


Asunto(s)
Ferroptosis/efectos de los fármacos , Rabdomiosarcoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclohexilaminas/metabolismo , Glutatión/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fenilendiaminas/metabolismo , Piperazinas/metabolismo , Pirazoles/farmacología , Pirazolonas , Piridinas/farmacología , Piridonas , Especies Reactivas de Oxígeno/metabolismo , Rabdomiosarcoma/metabolismo , alfa-Tocoferol/metabolismo
9.
Brain Res Bull ; 153: 122-132, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31442590

RESUMEN

Intracerebral hemorrhage (ICH) is a devastating subtype of stroke because it has few viable therapeutic options to intervene against primary or second brain injury. Recently, evidence has suggested that ferroptosis, a nonapoptotic form of cell death, is involved in ICH. In this study, we examined whether ICH-induced neuron death is partly ferroptotic in humans and assessed its temporal and spatial characteristics in mice. Furthermore, the ferroptosis inhibitor ferrostatin-1 (Fer-1) was used to examine the role of ferroptosis after ICH. Fold changes in ferroptosis-related gene expression, intracellular iron levels, malondialdehyde (MDA) levels, and both protein levels and cellular localization of cyclooxygenase-2 (COX-2) were measured to monitor ferroptosis. Transmission electron microscopy (TEM) was also performed to examine the ultrastructure of cells after ICH. We found that the expression level of prostaglandin-endoperoxide synthase (PTGS2) was increased in both in vitro and in vivo ICH models; by comparison, expression level of RPL8 was increased in human brain tissue. In mice, iron and MDA levels were significantly increased 3 h after ICH; COX-2 levels were increased at 12 h after ICH and peaked at 3 days after ICH; COX-2 colocalized with NeuN (a neuronal biomarker); and TEM showed that shrunken mitochondria were found at 3 h, 3 days, and 7 days after ICH. Moreover, ICH-induced neurological deficits, memory impairment and brain atrophy were reduced by Fer-1 treatment. Our results demonstrated that neuronal ferroptosis occurs during the acute phase of ICH in brain areas distant from the hematoma and that inhibition of ferroptosis by Fer-1 exerted a long-term cerebroprotective effect.


Asunto(s)
Hemorragia Cerebral/metabolismo , Ciclohexilaminas/farmacología , Ferroptosis/fisiología , Fenilendiaminas/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/fisiopatología , Ciclohexilaminas/metabolismo , Ciclooxigenasa 2/metabolismo , Humanos , Hierro/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Fenilendiaminas/metabolismo
10.
ACS Synth Biol ; 8(2): 346-357, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30586497

RESUMEN

Shinorine, a mycosporine-like amino acid (MAA), is a small molecule sunscreen produced in some bacteria. In this study, by introducing shinorine biosynthetic genes from cyanobacteria Nostoc punctiform into Saccharomyces cerevisiae, we successfully constructed yeast strains capable of producing shinorine. Sedoheptulose 7-phosphate (S7P), an intermediate of the pentose phosphate pathway, is a key substrate for shinorine biosynthesis. To increase the S7P pool, xylose, which is assimilated via the pentose phosphate pathway, was used as a carbon source after introducing xylose assimilation genes from Scheffersomyces stipitis into the shinorine-producing strain. The resulting xylose-fermenting strain produced a trace amount of shinorine when cells were grown in glucose, but shinorine production was dramatically increased by adding xylose in the medium. Shinorine production was further improved by modulating the pentose phosphate pathway through deleting TAL1 and overexpressing STB5 and TKL1. The final engineered strain JHYS17-4 produced 31.0 mg/L (9.62 mg/g DCW) of shinorine in the optimized medium containing 8 g/L of xylose and 12 g/L of glucose, demonstrating that S. cerevisiae is a promising host to produce this natural sunscreen material.


Asunto(s)
Ciclohexilaminas/metabolismo , Glicina/análogos & derivados , Ingeniería Metabólica/métodos , Saccharomyces cerevisiae/metabolismo , Xilosa/metabolismo , Glicina/metabolismo , Vía de Pentosa Fosfato , Fosfatos de Azúcar/metabolismo
11.
J Med Chem ; 61(22): 10126-10140, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30354101

RESUMEN

Ferroptosis is an iron-catalyzed, nonapoptotic form of regulated necrosis that results in oxidative lipid damage in cell membranes that can be inhibited by the radical-trapping antioxidant Ferrostatin-1 (Fer-1). Novel inhibitors derived from the Fer-1 scaffold inhibited ferroptosis potently but suffered from solubility issues. In this paper, we report the synthesis of a more stable and readily soluble series of Fer-1 analogues that potently inhibit ferroptosis. The most promising compounds (37, 38, and 39) showed an improved protection compared to Fer-1 against multiorgan injury in mice. No toxicity was observed in mice after daily injection of 39 (UAMC-3203) for 4 weeks. UAMC-3203 inserts rapidly in a phospholipid bilayer in silico, which aligns with the current understanding of the mechanism of action of these compounds. In conclusion, these analogues have superior properties compared to Fer-1, show in vivo efficacy, and represent novel lead compounds with therapeutic potential in relevant ferroptosis-driven disease models.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclohexilaminas/metabolismo , Diseño de Fármacos , Fenilendiaminas/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Humanos , Ratones , Modelos Moleculares , Conformación Molecular , Estrés Oxidativo/efectos de los fármacos , Ratas , Distribución Tisular
12.
Brain Res ; 1701: 112-125, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30205109

RESUMEN

Oxidative stress plays an important role in secondary brain injury (SBI) after intracerebral hemorrhage (ICH), but the underling mechanism has not been fully elucidated. Recently, the antioxidant enzyme glutathione peroxidase 4 (GPX4), has attracted increasing attention due to its ability to degrade reactive oxygen species (ROS) which are the major indicator of oxidative stress; However, the role of GPX4 in ICH has not been reported. This study was designed to investigate the changes in protein levels, as well as potential role and mechanism of GPX4 in SBI following ICH using a Sprague-Dawley (SD) rat model of ICH induced by autologous blood injection into the right basal ganglia. Firstly, GPX4 protein levels in the brain were reduced gradually and bottomed out at 24 h after ICH, compared with the Sham group. Secondly, genetic-overexpression of GPX4 effectively increased level of GPX4 in the brain, and clearly relieved neuronal dysfunction, brain edema, blood brain barrier (BBB) injury, oxidative stress and inflammation after ICH. In contrast, inhibiting GPX4 with a specific pharmacological inhibitor or genetic knockdown exacerbated SBI after ICH. Finally, Ferrostatin-1, a chemical inhibitor of ferroptosis, was used to explore the role of ferroptosis in brain injury after ICH. The results suggest that inhibiting ferroptosis can significantly alleviate SBI after ICH. In summary, our work indicated that GPX4 contributes to SBI following ICH by mediating ferroptosis. Therefore, inhibiting ferroptosis with specific inhibitors or upregulation of GPX4 may be a potential strategy to ameliorate brain injury induced by ICH.


Asunto(s)
Lesiones Encefálicas/enzimología , Hemorragia Cerebral/enzimología , Glutatión Peroxidasa/metabolismo , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Muerte Celular/fisiología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclohexilaminas/metabolismo , Masculino , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fenilendiaminas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
13.
J Gen Appl Microbiol ; 64(5): 203-211, 2018 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-29709901

RESUMEN

The aquatic cyanobacterium Nostoc verrucosum forms macroscopic colonies in streams, and its appearance is superficially similar to that of the terrestrial cyanobacterium Nostoc commune. N. verrucosum is sensitive to desiccation, unlike N. commune, although these Nostoc cyanobacterial species share physiological features, including massive extracellular polysaccharide production and trehalose accumulation capability. In this study, water-soluble sunscreen pigments of mycosporine-like amino acids (MAAs) were characterized in N. verrucosum, and the mysABCD genes responsible for MAA biosynthesis in N. verrucosum and N. commune were compared. N. verrucosum produced porphyra-334 and shinorine, with porphyra-334 accounting for >90% of the total MAAs. Interestingly, porphyra-334 is an atypical cyanobacteial MAA, whereas shinorine is known as a common and dominant MAA in cyanobacteria. Porphyra-334 from N. verrucosum showed little or no radical scavenging activity in vitro, although the glycosylated derivatives of porphyra-334 from N. commune are potent radical scavengers. The presence of the mysABCD gene cluster in N. commune strain KU002 (genotype A) supported its porphyra-334 producing capability via the Nostoc-type mechanism, although the genotype A of N. commune mainly produces the arabinose-bound porphyra-334. The mysABC gene cluster was conserved in N. verrucosum, but the mysD gene was not included in the cluster. These results suggest that the mysABCD gene products are involved in the biosynthesis of porphyra-334 commonly in these Nostoc species, and that the genotype A of N. commune additionally acquired the glycosylation of porphyra-334.


Asunto(s)
Ciclohexanonas , Ciclohexilaminas , Glicina/análogos & derivados , Nostoc/química , Ciclohexanonas/metabolismo , Ciclohexilaminas/metabolismo , Glicina/biosíntesis , Glicina/genética , Glicina/metabolismo , Glicosilación , Familia de Multigenes/genética , Nostoc/genética , Protectores Solares/química
14.
Elife ; 72018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29489452

RESUMEN

eIF2B is a dedicated guanine nucleotide exchange factor for eIF2, the GTPase that is essential to initiate mRNA translation. The integrated stress response (ISR) signaling pathway inhibits eIF2B activity, attenuates global protein synthesis and upregulates a set of stress-response proteins. Partial loss-of-function mutations in eIF2B cause a neurodegenerative disorder called Vanishing White Matter Disease (VWMD). Previously, we showed that the small molecule ISRIB is a specific activator of eIF2B (Sidrauski et al., 2015). Here, we report that various VWMD mutations destabilize the decameric eIF2B holoenzyme and impair its enzymatic activity. ISRIB stabilizes VWMD mutant eIF2B in the decameric form and restores the residual catalytic activity to wild-type levels. Moreover, ISRIB blocks activation of the ISR in cells carrying these mutations. As such, ISRIB promises to be an invaluable tool in proof-of-concept studies aiming to ameliorate defects resulting from inappropriate or pathological activation of the ISR.


Asunto(s)
Acetamidas/metabolismo , Ciclohexilaminas/metabolismo , Activadores de Enzimas/metabolismo , Factor 2B Eucariótico de Iniciación/metabolismo , Leucoencefalopatías/fisiopatología , Proteínas Mutantes/metabolismo , Fármacos Neuroprotectores/metabolismo , Línea Celular , Factor 2B Eucariótico de Iniciación/genética , Humanos , Leucoencefalopatías/genética , Proteínas Mutantes/genética
15.
Redox Biol ; 15: 394-404, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29331741

RESUMEN

The extracellular pH (pHe) is a key determinant of the cellular (micro)environment and needs to be maintained within strict boundaries to allow normal cell function. Here we used HEK293 cells to study the effects of pHe acidification (24h), induced by mitochondrial inhibitors (rotenone, antimycin A) and/or extracellular HCl addition. Lowering pHe from 7.2 to 5.8 reduced cell viability by 70% and was paralleled by a decrease in cytosolic pH (pHc), hyperpolarization of the mitochondrial membrane potential (Δψ), increased levels of hydroethidine-oxidizing ROS and stimulation of protein carbonylation. Co-treatment with the antioxidant α-tocopherol, the mitochondrial permeability transition pore (mPTP) desensitizer cyclosporin A and Necrostatin-1, a combined inhibitor of Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and Indoleamine 2,3-dioxygenase (IDO), prevented acidification-induced cell death. In contrast, the caspase inhibitor zVAD.fmk and the ferroptosis inhibitor Ferrostatin-1 were ineffective. We conclude that extracellular acidification induces necroptotic cell death in HEK293 cells and that the latter involves intracellular acidification, mitochondrial functional impairment, increased ROS levels, mPTP opening and protein carbonylation. These findings suggest that acidosis of the extracellular environment (as observed in mitochondrial disorders, ischemia, acute inflammation and cancer) can induce cell death via a ROS- and mPTP opening-mediated pathogenic mechanism.


Asunto(s)
Ácidos/metabolismo , Microambiente Celular/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ácidos/farmacología , Animales , Antimicina A/farmacología , Supervivencia Celular/efectos de los fármacos , Ciclohexilaminas/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Fenilendiaminas/metabolismo , Carbonilación Proteica/efectos de los fármacos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Rotenona/farmacología
16.
Arch Toxicol ; 92(2): 759-775, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28975372

RESUMEN

Reactive oxygen species (ROS)-induced apoptosis has been extensively studied. Increasing evidence suggests that ROS, for instance, induced by hydrogen peroxide (H2O2), might also trigger regulated necrotic cell death pathways. Almost nothing is known about the cell death pathways triggered by tertiary-butyl hydroperoxide (t-BuOOH), a widely used inducer of oxidative stress. The lipid peroxidation products induced by t-BuOOH are involved in the pathophysiology of many diseases, such as cancer, cardiovascular diseases, or diabetes. In this study, we exposed murine fibroblasts (NIH3T3) or human keratinocytes (HaCaT) to t-BuOOH (50 or 200 µM, respectively) which induced a rapid necrotic cell death. Well-established regulators of cell death, i.e., p53, poly(ADP)ribose polymerase-1 (PARP-1), the stress kinases p38 and c-Jun N-terminal-kinases 1/2 (JNK1/2), or receptor-interacting serine/threonine protein kinase 1 (RIPK1) and 3 (RIPK3), were not required for t-BuOOH-mediated cell death. Using the selective inhibitors ferrostatin-1 (1 µM) and liproxstatin-1 (1 µM), we identified ferroptosis, a recently discovered cell death mechanism dependent on iron and lipid peroxidation, as the main cell death pathway. Accordingly, t-BuOOH exposure resulted in a ferrostatin-1- and liproxstatin-1-sensitive increase in lipid peroxidation and cytosolic ROS. Ferroptosis was executed independently from other t-BuOOH-mediated cellular damages, i.e., loss of mitochondrial membrane potential, DNA double-strand breaks, or replication block. H2O2 did not cause ferroptosis at equitoxic concentrations (300 µM) and induced a (1) lower and (2) ferrostatin-1- or liproxstatin-1-insensitive increase in lipid peroxidation. We identify that t-BuOOH and H2O2 produce a different pattern of lipid peroxidation, thereby leading to different cell death pathways and present t-BuOOH as a novel inducer of ferroptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Peroxidación de Lípido , terc-Butilhidroperóxido/toxicidad , Animales , Cardiolipinas/metabolismo , Línea Celular , Supervivencia Celular , Ciclohexilaminas/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Queratinocitos/citología , Potencial de la Membrana Mitocondrial , Ratones , Células 3T3 NIH , Fenilendiaminas/metabolismo , Quinoxalinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Espiro/metabolismo
17.
Curr Microbiol ; 75(3): 284-287, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29063968

RESUMEN

Acinetobacter sp. YT-02, a Gram-negative bacterium isolated from the activated sludge from a sodium N-cyclohexylsulfamate production plant, has the ability to degrade cyclohexylamine. It was classified as a member of Acinetobacter sp., a Gram-negative bacterium, sharing a 16S rRNA gene sequence identity of 99% with Acinetobacter guangdongensis strain 1NM-4. It could degrade 10 mmol/L cyclohexylamine within 22 h. Based on the identified metabolite, the metabolic pathway of cyclohexylamine could be postulated as it was degraded via cyclohexanone. Draft genome sequence of this strain (2,993, 647 bp of chromosome length) is presented here. We further identified the genes encoding the enzymes involved in cyclohexylamine oxidation to cyclohexanone and the subsequent downstream metabolic pathway of cyclohexanone oxidation. Strain YT-02 has the potentiality to be applied in the treatment of the pollutant cyclohexylamine, and it could also be treated as a research material to study the degradation mechanism of cyclohexylamine.


Asunto(s)
Acinetobacter/genética , Acinetobacter/aislamiento & purificación , Ciclohexilaminas/metabolismo , Genoma Bacteriano , Acinetobacter/clasificación , Acinetobacter/metabolismo , Secuencia de Bases , Biodegradación Ambiental , ADN Bacteriano/genética , Datos de Secuencia Molecular , Filogenia , ARN Ribosómico 16S/genética , Aguas del Alcantarillado/microbiología
18.
Chemistry ; 23(51): 12604-12612, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28661578

RESUMEN

The glmS ribozyme is a bacterial gene-regulating riboswitch that controls cell wall synthesis, depending on glucosamine-6-phosphate as a cofactor. Due to the presence of this ribozyme in several human pathogen bacteria (e.g., MRSA, VRSA), the glmS ribozyme represents an attractive target for the development of artificial cofactors. The substitution of the ring oxygen in carbohydrates by functionalized methylene groups leads to a new generation of glycomimetics that exploits distinct interaction possibilities with their target structure in biological systems. Herein, we describe the synthesis of mono-fluoro-modified carba variants of α-d-glucosamine and ß-l-idosamine. (5aR)-Fluoro-carba-α-d-glucosamine-6-phosphate is a synthetic mimic of the natural ligand of the glmS ribozyme and is capable of effectively addressing its unique self-cleavage mechanism. However, in contrast to what was expected, the activity is significantly decreased compared to its non-fluorinated analog. By combining self-cleavage assays with the Bacillus subtilis and Staphylococcus aureus glmS ribozyme and molecular docking studies, we provide a structure-activity relationship for fluorinated carba-sugars.


Asunto(s)
Proteínas Bacterianas/metabolismo , Carba-azúcares/metabolismo , ARN Catalítico/metabolismo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/química , Sitios de Unión , Carba-azúcares/síntesis química , Carba-azúcares/química , Ciclohexanoles/síntesis química , Ciclohexanoles/química , Ciclohexanoles/metabolismo , Ciclohexilaminas/síntesis química , Ciclohexilaminas/química , Ciclohexilaminas/metabolismo , Halogenación , Conformación Molecular , Simulación del Acoplamiento Molecular , Conformación de Ácido Nucleico , ARN Catalítico/química , Staphylococcus aureus/metabolismo , Estereoisomerismo , Relación Estructura-Actividad
19.
J Biotechnol ; 251: 166-173, 2017 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-28472672

RESUMEN

The Gram-negative strain of Pseudomonas plecoglossicida NyZ12 isolated from soil has the ability to degrade cyclohexylamine (CHAM). The genes encoding CHAM degradation by gram-negative bacteria, however, have not been reported previously. In this study, ORFs predicted to encode CHAM degradation by NyZ12 were identified by bioinformatics analysis. Differential expression of the proposed ORFs was analyzed via RNA-seq and quantitative reverse transcription-PCR (qRT-PCR), using RNA extracted from NyZ12 cultured with or without CHAM addition. One CHAM-inducible ORF, RK21_02867 predicted to encode a cyclohexanone monooxygenase (ChnB) was disrupted, as were five ORFs, RK21_00425, RK21_02631, RK21_04207, RK21_04637 and RK21_05539, that had weak homology to the only known cyclohexylamine oxidase (CHAO encoded by chaA) found in Brevibacterium oxydans IH-35A. We also found that a tandem array of five ORFs (RK21_02866-02870) shared homology with those in an operon responsible for oxidation of cyclohexanone to adipic acid, although the ORFs in strain NyZ12 were arranged in a different order with previously found in cyclohexane, cyclohexanol or cyclohexanone degradation strains. The ORFs in this cluster were all up-regulated when CHAM was supplied as the sole carbon source. When one of these five genes, RK21_02867 encoding cyclohexanone (CHnone) monooxygenase, was knocked out, NyZ12 could not grow on CHAM, but it accumulated equimolar amounts of CHnone. Our results show that strain NyZ12 metabolized CHAM directly to CHnone which was then further metabolized to adipate. Despite clearly identifying genes encoding the steps for metabolism of CHAM metabolites, not every one of the putative chaAs was differentially expressed in the presence of CHAM and deletion of each one individually did not completely eliminate the capacity of NyZ12 to degrade CHAM, though it did reduce its growth in several instances. Our results suggest that there is genetic redundancy encoding the initial step in the oxidation of CHAM to CHnone in NyZ12 and that its CHAOs differ considerably from the ChaA, originally described in Brevibacterium oxydans IH-35A.


Asunto(s)
Genoma Bacteriano , Pseudomonas/genética , Biodegradación Ambiental , Ciclohexilaminas/metabolismo , Genes Bacterianos , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Pseudomonas/metabolismo
20.
Curr Top Microbiol Immunol ; 403: 143-170, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28204974

RESUMEN

Glutathione peroxidase 4 (Phospholipid hydroperoxide glutathione peroxidase, PHGPx) can directly reduce phospholipid hydroperoxide. Depletion of GPx4 induces lipid peroxidation-dependent cell death in embryo, testis, brain, liver, heart, and photoreceptor cells of mice. Administration of vitamin E in tissue specific GPx4 KO mice restored tissue damage in testis, liver, and heart. These results indicate that suppression of phospholipid peroxidation is essential for cell survival in normal tissues in mice. Ferroptosis is an iron-dependent non-apoptotic cell death that can elicited by pharmacological inhibiting the cystine/glutamate antiporter, system Xc- (type I) or directly binding and loss of activity of GPx4 (Type II) in cancer cells with high level RAS-RAF-MEK pathway activity or p53 expression, but not in normal cells. Ferroptosis by Erastin (Type I) and RSL3 (RAS-selective lethal 3, Type II) treatment was suppressed by an iron chelator, vitamin E and Ferrostatin-1, antioxidant compound. GPx4 can regulate ferroptosis by suppression of phospholipid peroxidation in erastin and RSL3-induced ferroptosis. Recent works have identified several regulatory factors of erastin and RSL3-induced ferroptosis. In our established GPx4-deficient MEF cells, depletion of GPx4 induce iron and 15LOX-independent lipid peroxidation at 26 h and caspase-independent cell death at 72 h, whereas erastin and RSL3 treatment resulted in iron-dependent ferroptosis by 12 h. These results indicated the possibility that the mechanism of GPx4-depleted cell death might be different from that of ferroptosis induced by erastin and RSL3.


Asunto(s)
Muerte Celular/fisiología , Ciclohexilaminas/metabolismo , Glutatión Peroxidasa/metabolismo , Hierro/metabolismo , Peroxidación de Lípido/fisiología , Fenilendiaminas/metabolismo , Animales , Carbolinas/farmacología , Caspasas/metabolismo , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Piperazinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...