Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
J Inorg Biochem ; 244: 112238, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37119547

RESUMEN

Human heme oxygenase-1 (hHO-1) plays a crucial role in human physiology because of its ability to metabolize free heme. The heme degradation products, biliverdin and bilirubin, were shown to have protective antioxidant properties in cells. In the context of cancer, hHO-1 function grants cancer cells defense from standard chemotherapy treatments, leading to the development of azole-based inhibitors that target hHO-1 for potential anticancer therapy. This work reports experimental and theoretical characterization of interactions between three azole-based inhibitors and the active site of hHO-1. It was found that all three compounds have Kd values within the µM order. The electronic absorption and resonance Raman (rR) spectra indicated that they bind to the ferric heme and coordinate through a nitrogen atom. rR measurements revealed varying effects of inhibitors on the geometry of heme vinyl groups in the ferric form of hHO-1. Changes in peripheral group orientation are known to affect heme redox potential, and consequently can reflect the inhibitory properties of studied azoles. The subsequent docking studies showed that inhibitors with lower Kd values are located close to two vinyl groups, while the compound with higher Kd is situated near only one, consistent with the rR studies. Finally, the rR studies of the CO adducts showed that the inhibitors bind to the heme in a reversible manner. Altogether, the combination of ligand binding studies, UV-Vis and rR spectroscopies, as well as computational approach revealed an importance of the steric hindrance imposed by the inhibitor's side chain.


Asunto(s)
Azoles , Hemo Oxigenasa (Desciclizante) , Humanos , Hemo Oxigenasa (Desciclizante)/química , Azoles/farmacología , Hemo-Oxigenasa 1/química , Espectrometría Raman , Hierro , Hemo/química
2.
Cell Death Dis ; 12(8): 738, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34312365

RESUMEN

Heme oxygenase-1 (HO-1) has attracted accumulating attention for its antioxidant enzymatic activity. However, the exact regulatory role of its non-enzymatic activity in the cardiovascular system remains unaddressed. Here, we show that HO-1 was accumulated in the nuclei of stress-induced senescent endothelial cells, and conferred protection against endothelial senescence independent of its enzymatic activity. Overexpression of ΔHO-1, a truncated HO-1 without transmembrane segment (TMS), inhibited H2O2-induced endothelial senescence. Overexpression of ΔHO-1H25A, the catalytically inactive form of ΔHO-1, also exhibited anti-senescent effect. In addition, infection of recombinant adenovirus encoding ΔHO-1 with three nuclear localization sequences (NLS), alleviated endothelial senescence induced by knockdown of endogenous HO-1 by CRISPR/Cas9. Moreover, repression of HO-1 nuclear translocation by silencing of signal peptide peptidase (SPP), which is responsible for enzymatic cleavage of the TMS of HO-1, exacerbated endothelial senescence. Mechanistically, nuclear HO-1 interacted with NPM1 N-terminal portion, prevented NPM1 translocation from nucleolus to nucleoplasm, thus disrupted NPM1/p53/MDM2 interactions and inhibited p53 activation by NPM1, finally resisted endothelial senescence. This study provides a novel understanding of HO-1 as a promising therapeutic strategy for vascular senescence-related cardiovascular diseases.


Asunto(s)
Núcleo Celular/metabolismo , Senescencia Celular , Hemo-Oxigenasa 1/metabolismo , Nucleofosmina/metabolismo , Estrés Fisiológico , Envejecimiento/genética , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Senescencia Celular/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Mutación/genética , Nucleofosmina/química , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
3.
J Enzyme Inhib Med Chem ; 36(1): 1378-1386, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34167427

RESUMEN

In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Predicted in silico physicochemical properties of the newly synthesised hybrid (3) demonstrated a drug-like profile with suitable Absorption, Distribution, Metabolism, and Excretion (ADME) properties and low toxic liabilities. Preliminary cytotoxicity evaluation towards human prostate (DU145) and lung (A549) cancer cell lines demonstrated that 3 exerted a similar effect on cell viability to that produced by the reference drug 5-FU. Among the two tested cancer cell lines, the A549 cells were more susceptible for 3. Of note, hybrid 3 also had a significantly lower cytotoxic effect on healthy human lung epithelial cells (BEAS-2B) than 5-FU. Altogether our results served as an initial proof-of-concept to develop 5-FU/HO-1 mutual prodrugs as potential novel anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Fluorouracilo/química , Hemo-Oxigenasa 1/química , Profármacos/química , Profármacos/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Profármacos/síntesis química , Ratas , Ratas Sprague-Dawley , Porcinos
4.
J Biol Chem ; 296: 100217, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33839679

RESUMEN

Heme oxygenase (HO) converts heme to carbon monoxide, biliverdin, and free iron, products that are essential in cellular redox signaling and iron recycling. In higher plants, HO is also involved in the biosynthesis of photoreceptor pigment precursors. Despite many common enzymatic reactions, the amino acid sequence identity between plant-type and other HOs is exceptionally low (∼19.5%), and amino acids that are catalytically important in mammalian HO are not conserved in plant-type HOs. Structural characterization of plant-type HO is limited to spectroscopic characterization by electron spin resonance, and it remains unclear how the structure of plant-type HO differs from that of other HOs. Here, we have solved the crystal structure of Glycine max (soybean) HO-1 (GmHO-1) at a resolution of 1.06 Å and carried out the isothermal titration calorimetry measurements and NMR spectroscopic studies of its interaction with ferredoxin, the plant-specific electron donor. The high-resolution X-ray structure of GmHO-1 reveals several novel structural components: an additional irregularly structured region, a new water tunnel from the active site to the surface, and a hydrogen-bonding network unique to plant-type HOs. Structurally important features in other HOs, such as His ligation to the bound heme, are conserved in GmHO-1. Based on combined data from X-ray crystallography, isothermal titration calorimetry, and NMR measurements, we propose the evolutionary fine-tuning of plant-type HOs for ferredoxin dependency in order to allow adaptation to dynamic pH changes on the stroma side of the thylakoid membrane in chloroplast without losing enzymatic activity under conditions of fluctuating light.


Asunto(s)
Ferredoxinas/química , Glycine max/química , Hemo-Oxigenasa 1/química , Hemo/química , Hierro/química , Proteínas de Plantas/química , Secuencia de Aminoácidos , Biliverdina/química , Biliverdina/metabolismo , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Dominio Catalítico , Cloroplastos/química , Cloroplastos/enzimología , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Ferredoxinas/genética , Ferredoxinas/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Hemo/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Enlace de Hidrógeno , Hierro/metabolismo , Simulación del Acoplamiento Molecular , Resonancia Magnética Nuclear Biomolecular , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Glycine max/enzimología , Glycine max/genética , Tilacoides/química , Tilacoides/enzimología
5.
J Am Chem Soc ; 143(17): 6460-6469, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33845576

RESUMEN

Heme oxygenase-1 (HO-1) is a vital enzyme in humans that primarily regulates free heme concentrations. The overexpression of HO-1 is commonly associated with cardiovascular and neurodegenerative diseases including atherosclerosis and ischemic stroke. Currently, there are no known chemical probes to detect HO-1 activity, limiting its potential as an early diagnostic/prognostic marker in these serious diseases. Reported here are the design, synthesis, and photophysical and biological characterization of a coumarin-porphyrin FRET break-apart probe to detect HO-1 activity, Fe-L1. We designed Fe-L1 to "break-apart" upon HO-1-catalyzed porphyrin degradation, perturbing the efficient FRET mechanism from a coumarin donor to a porphyrin acceptor fluorophore. Analysis of HO-1 activity using Escherichia coli lysates overexpressing hHO-1 found that a 6-fold increase in emission intensity at 383 nm was observed following incubation with NADPH. The identities of the degradation products following catabolism were confirmed by MALDI-MS and LC-MS, showing that porphyrin catabolism was regioselective at the α-position. Finally, through the analysis of Fe-L2, we have shown that close structural analogues of heme are required to maintain HO-1 activity. It is anticipated that this work will act as a foundation to design and develop new probes for HO-1 activity in the future, moving toward applications of live fluorescent imaging.


Asunto(s)
Cumarinas/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Hemo-Oxigenasa 1/química , Protoporfirinas/química , Escherichia coli/enzimología , Escherichia coli/genética , Hemo-Oxigenasa 1/análisis , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Procesos Fotoquímicos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrofotometría Ultravioleta
6.
Protein J ; 40(2): 166-174, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33646477

RESUMEN

Upregulation of Heme Oxygenase-1 (HO-1) has been widely implicated in cancer growth and chemoresistance. This explains the numerous drug discovery efforts aimed at mitigating its pro-carcinogenic roles till date. In a recent study, two selective azole-based HO-1 inhibitors (Cpd1 and Cpd2) were synthesized, which exhibited differential inhibitory potencies of ~200µm. Interestingly, variations in the affinities of these compounds were determined by their positioning across specific regions of the HO-1 binding domain, pin-pointing a pharmacological interrelationship that remains unresolved. Therefore, in this study, using molecular dynamics simulations and binding free energy calculations, we investigate how dynamical orientations of these compounds influence their binding affinities at the active HO-1 domain. Findings revealed favorable binding for the bromobenzene and imidazole substituents of Cpd1 at the western and eastern regions of the HO-1 active domain. The constituent hydroxyl group was coordinated by residues Asp140 and Arg136 over the simulation period. On the contrary, stable binding of the bromobenzene and imidazole substituents were negated by the optimal orientations of the benzyl substituent, which extended into the northeastern region. These were supported by the displacement of Asp140 and Arg136, crucial for hydrogen bond formation in Cpd1. Also, we observed that Cpd2 exhibited high deviations indicative of an unstable binding relative to Cpd1. This further supports the presumption that Cpd2 was systematically oriented away from the active HO-1 region, a phenomenon that was due to the optimal motions of the benzyl group at the northeastern regions. The highlight of our findings is that the benzyl substituent in Cpd2 elicited negative effects on HO-1, vis a vis, instability, displacement of crucial residues, and low binding energy when compared to Cpd1. Findings pave the way for future drug discovery efforts related to HO-1 inhibition in cancer therapy.


Asunto(s)
Antineoplásicos , Inhibidores Enzimáticos , Hemo-Oxigenasa 1 , Imidazoles , Antineoplásicos/química , Antineoplásicos/metabolismo , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/metabolismo , Humanos , Imidazoles/química , Imidazoles/metabolismo , Simulación de Dinámica Molecular , Termodinámica
7.
Biochem J ; 478(2): 377-388, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33394027

RESUMEN

P450 and heme oxygenase-1 (HO-1) receive their necessary electrons by interaction with the NADPH-cytochrome P450 reductase (POR). As the POR concentration is limiting when compared with P450 and HO-1, they must effectively compete for POR to function. In addition to these functionally required protein-protein interactions, HO-1 forms homomeric complexes, and several P450s have been shown to form complexes with themselves and with other P450s, raising the question, 'How are the HO-1 and P450 systems organized in the endoplasmic reticulum?' Recently, CYP1A2 was shown to associate with HO-1 affecting the function of both proteins. The goal of this study was to determine if CYP1A1 formed complexes with HO-1 in a similar manner. Complex formation among POR, HO-1, and CYP1A1 was measured using bioluminescence resonance energy transfer, with results showing HO-1 and CYP1A1 form a stable complex that was further stabilized in the presence of POR. The POR•CYP1A1 complex was readily disrupted by the addition of HO-1. CYP1A1 also was able to affect the POR•HO-1 complex, although the effect was smaller. This interaction between CYP1A1 and HO-1 also affected function, where the presence of CYP1A1 inhibited HO-1-mediated bilirubin formation by increasing the KmPOR•HO-1 without affecting the Vmaxapp. In like manner, HO-1 inhibited CYP1A1-mediated 7-ethoxyresorufin dealkylation by increasing the KmPOR•CYP1A1. Based on the mathematical simulation, the results could not be explained by a model where CYP1A1 and HO-1 simply compete for POR, and are consistent with the formation of a stable CYP1A1•HO-1 complex that affected the functional characteristics of both moieties.


Asunto(s)
Citocromo P-450 CYP1A1/metabolismo , Hemo-Oxigenasa 1/metabolismo , Transferencia de Energía por Resonancia de Bioluminiscencia , Citocromo P-450 CYP1A1/química , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Hemo-Oxigenasa 1/química , Humanos , Dominios y Motivos de Interacción de Proteínas
8.
Fish Shellfish Immunol ; 112: 168-178, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32927052

RESUMEN

Heme oxygenase (HO)-1, a rate-limiting enzyme in heme catabolism, results in the formation of equivalent amounts of biliverdin (BV), carbon monoxide (CO) and ferrous iron (Fe2+). Previous studies have revealed that HO-1 plays an important role in immune responses. However, the mechanism underlying the immune responses against bacterial infection of teleost HO-1 remains enigmatic. To decipher the mechanisms, we have cloned and characterized the HO-1 gene of grass carp (designated as GcHO-1) in this research. The results showed that the open reading frame (ORF) of GcHO-1 was 819 bp, which encoded a putative protein of 272 amino acids. The deduced amino acid sequence phylogenetically shared the highest identity with other teleosts, and contained two domains of heme-oxygenase and a single-pass transmembrane domain. The mRNA expressions of GcHO-1 in healthy grass carp have widely existed in examined tissues in the following order of spleen > head-kidney > middle head-kidney > intestines > liver > gills > heart > muscle > brain. Besides, the mRNA and protein transcription of GcHO-1 were both significantly up-regulated in the liver and head-kidney tissues after Staphylococcus aureus and Aeromonas hydrophila infection. In addition, overexpression of GcHO-1 in kidney cell line (CIK) cells of grass carp could reduce the expression of inflammatory cytokines (IL-1ß, IL-8, TNFα, CCL1 and IL-6). Herein, we demonstrate that GcHO-1 plays an anti-inflammatory role in innate immunity. Our results shed new light on the mechanisms underlying the antibacterial immunity of teleost.


Asunto(s)
Carpas/genética , Carpas/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Inmunidad Innata/genética , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Enfermedades de los Peces/microbiología , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Hemo-Oxigenasa 1/química , Filogenia , Alineación de Secuencia/veterinaria , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/veterinaria , Staphylococcus aureus/fisiología
9.
J Biol Chem ; 294(20): 8259-8272, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30944174

RESUMEN

Heme oxygenase (HO) catalyzes heme degradation, a process crucial for regulating cellular levels of this vital, but cytotoxic, cofactor. Two HO isoforms, HO1 and HO2, exhibit similar catalytic mechanisms and efficiencies. They also share catalytic core structures, including the heme-binding site. Outside their catalytic cores are two regions unique to HO2: a 20-amino acid-long N-terminal extension and a C-terminal domain containing two heme regulatory motifs (HRMs) that bind heme independently of the core. Both HO isoforms contain a C-terminal hydrophobic membrane anchor; however, their sequences diverge. Here, using hydrogen-deuterium exchange MS, size-exclusion chromatography, and sedimentation velocity, we investigated how these divergent regions impact the dynamics and structure of the apo and heme-bound forms of HO1 and HO2. Our results reveal that heme binding to the catalytic cores of HO1 and HO2 causes similar dynamic and structural changes in regions (proximal, distal, and A6 helices) within and linked to the heme pocket. We observed that full-length HO2 is more dynamic than truncated forms lacking the membrane-anchoring region, despite sharing the same steady-state activity and heme-binding properties. In contrast, the membrane anchor of HO1 did not influence its dynamics. Furthermore, although residues within the HRM domain facilitated HO2 dimerization, neither the HRM region nor the N-terminal extension appeared to affect HO2 dynamics. In summary, our results highlight significant dynamic and structural differences between HO2 and HO1 and indicate that their dissimilar C-terminal regions play a major role in controlling the structural dynamics of these two proteins.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/química , Hemo-Oxigenasa 1/química , Hemo/química , Simulación de Dinámica Molecular , Multimerización de Proteína , Secuencias de Aminoácidos , Medición de Intercambio de Deuterio , Hemo/genética , Hemo/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Dominios Proteicos
10.
ChemMedChem ; 13(13): 1336-1342, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29693778

RESUMEN

A 3D quantitative structure-activity relationship (3D-QSAR) model for predicting the activity of heme oxygenase 1 (HO-1) inhibitors was constructed with the aim of providing a useful tool for the identification, design, and optimization of novel HO-1 inhibitors. The model was built using a set of 222 HO-1 inhibitors recovered from the Heme Oxygenase Database (HemeOxDB) and developed with the software Forge. The present model showed high statistical quality, as confirmed by its robust predictive potential and satisfactory descriptive capability. The drawn-up 3D map enables prompt visual comprehension of the electrostatic, hydrophobic, and shaping features underlying the interactions with HO-1 inhibitors. A theoretical approach for the generation of new lead compounds was performed by means of scaffold-hopping analysis. For the first time, a 3D-QSAR model is reported for this target, and was built with a number of chemically diverse HO-1 inhibitors; the model also accounts well for individual ligand affinities. The new model contains all of the inhibitors published to date with high potency toward the selected target and contains a complete pharmacophore description of the binding cavity of HO-1. These features will ensure application in accelerating the identification of new potent and selective HO-1 inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/antagonistas & inhibidores , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad Cuantitativa , Análisis de Regresión , Programas Informáticos
11.
Biochem Biophys Res Commun ; 500(2): 429-434, 2018 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-29660345

RESUMEN

Alternative splicing is a routine phenomenon which greatly increases the diversity of proteins in eukaryotic cells. In humans, most multi-exonic genes are alternatively spliced and their splice variants confer distinct functions. Heme oxygenase-1 (HO-1, 32 kDa) is an inducible stress responsive protein, which possesses multiple functions in many cellular processes. In the current study, we identified a novel alternative splice isoform of 14 kDa HO-1 generated through exclusion of exon 3, and it is highly expressed in immortalized cells. In contrast to nuclear accumulation of the full-length 32 kDa HO-1, the novel 14 kDa HO-1 isoform is retained in the cytoplasm under ultraviolet (UV) irradiation. Interestingly, the 14 kDa HO-1 is shown to promote cell proliferation and an increase in relative telomere lengths in vivo and in vitro. Thus, we are pioneer to report and confirm the presence of a novel splice form of HO-1 and its distinct role in modulating telomere length and tumor growth.


Asunto(s)
Empalme Alternativo/genética , Hemo-Oxigenasa 1/genética , Telómero/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proliferación Celular , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Peso Molecular , Transporte de Proteínas , Fracciones Subcelulares/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-29698685

RESUMEN

Cadmium is a highly toxic environmental pollutant that can cause many adverse effects including cancer, neurological disease and kidney damage. Aquatic amphibians are particularly susceptible to this toxicant as it was shown to cause developmental abnormalities and genotoxic effects. In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. In the present study, CdCl2 treatment of A6 kidney epithelial cells, derived from the frog, Xenopus laevis, induced the accumulation of HO-1, heat shock protein 70 (HSP70) and HSP30 as well as an increase in the production of aggregated protein and aggresome-like structures. Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. Treatment of cells with hemin and baicalein, which were previously shown to provide cytoprotection against various stresses, induced HO-1 accumulation in a concentration-dependent manner. Also, treatment of cells with hemin and baicalein suppressed CdCl2-induced actin dysregulation and the accumulation of aggregated protein and aggresome-like structures. This cytoprotective effect was inhibited by SnPP. These results suggest that HO-1-mediated protection against CdCl2 toxicity includes the maintenance of actin cytoskeletal and microtubular structure and the suppression of aggregated protein and aggresome-like structures.


Asunto(s)
Cadmio/toxicidad , Contaminantes Ambientales/toxicidad , Proteínas del Choque Térmico HSP30/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Hemo-Oxigenasa 1/metabolismo , Riñón/efectos de los fármacos , Agregación Patológica de Proteínas/inducido químicamente , Animales , Antioxidantes/química , Antioxidantes/metabolismo , Línea Celular , Suplementos Dietéticos , Inhibidores Enzimáticos/farmacología , Flavanonas/antagonistas & inhibidores , Flavanonas/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/química , Hemina/antagonistas & inhibidores , Hemina/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/patología , Metaloporfirinas/farmacología , Microscopía Confocal , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/prevención & control , Protoporfirinas/farmacología , Proteínas de Xenopus/agonistas , Proteínas de Xenopus/antagonistas & inhibidores , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis
13.
Phytother Res ; 32(5): 892-897, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29356228

RESUMEN

To elucidate the anti-inflammatory mechanism of Curcuma sp., we investigated whether dehydrocurdione, a sesquiterpene contained in Curcuma sp., induces heme oxygenase (HO)-1, an antioxidative enzyme, in RAW 264.7 macrophages. Dehydrocurdione was extracted from the rhizome of Curcuma sp., and its purity was verified by high performance liquid chromatography. Treatment with 10-100 µM dehydrocurdione transiently and concentration-dependently increased HO-1 mRNA and protein levels. Docking simulation suggested the presence of the Michael reaction between dehydrocurdione and Kelch-like ECH-associated protein (Keap)1 keeping nuclear factor-erythroid2-related-factor (Nrf)2, a transcription factor, in the cytoplasm. Nrf2 that was definitely free from Keap1 was detected in the nuclei after dehydrocurdione treatment. Subsequently, the HO-1 E2 enhancer, a target of Nrf2, was activated, resulting in HO-1 expression. Also, an investigation using 6-shogaol and 6-gingerol supported the concept that the α, ß-unsaturated carbonyl structure plays an important role in the interaction with Keap1. Dehydrocurdione suppressed lipopolysaccharide-induced NO release, a marker of inflammation. Clarification of the HO-1 synthesis increase mechanism revealed in this study will help contribute to the development of novel phytotherapeutic strategies against inflammation-associated diseases.


Asunto(s)
Antiinflamatorios/farmacología , Curcuma/química , Hemo-Oxigenasa 1/metabolismo , Sesquiterpenos/farmacología , Animales , Antiinflamatorios/aislamiento & purificación , Antioxidantes/farmacología , Células Cultivadas , Inducción Enzimática/efectos de los fármacos , Células HEK293 , Hemo-Oxigenasa 1/química , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Extractos Vegetales/química , Extractos Vegetales/farmacología , Unión Proteica/efectos de los fármacos , Carbonilación Proteica , Dominios y Motivos de Interacción de Proteínas , Sesquiterpenos/aislamiento & purificación
14.
J Biochem Mol Toxicol ; 32(1)2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29140578

RESUMEN

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates a cluster of oxidative stress-inducible genes in cells. Here, we aimed to investigate whether trehalose (Tre) protects primary rat proximal tubular (rPT) cells against cadmium (Cd)-induced oxidative stress via Nrf2 antioxidant pathway. Data showed that Tre treatment inhibited Nrf2 nuclear translocation and restored the decline in Kelch-like ECH-associated protein 1 (Keap1) protein level in Cd-exposed rPT cells. Moreover, Cd-activated Nrf2 target genes, including phase II detoxifying enzymes, that is, NAD(P)H quinone oxidoreductase 1 and heme oxygenase-1, direct antioxidant proteins, that is, glutathione peroxidase, superoxide dismutase, catalase, and glutathione biosynthesis-related proteins, that is, glutamatecysteine ligase catalytic subunit, glutamate cysteine ligase modifier subunit, and glutathione reductase, were all downregulated by co-treatment with Tre. Collectively, these findings demonstrate that Tre treatment alleviates Cd-induced oxidative stress in rPT cells by inhibiting the Nrf2-Keap1 signaling pathway.


Asunto(s)
Cadmio/toxicidad , Proteína 1 Asociada A ECH Tipo Kelch/antagonistas & inhibidores , Túbulos Renales Proximales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trehalosa/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Cadmio/química , Intoxicación por Cadmio/dietoterapia , Intoxicación por Cadmio/metabolismo , Intoxicación por Cadmio/patología , Intoxicación por Cadmio/prevención & control , Catalasa/antagonistas & inhibidores , Catalasa/química , Catalasa/metabolismo , Células Cultivadas , Suplementos Dietéticos , Regulación hacia Abajo , Glutatión Reductasa/antagonistas & inhibidores , Glutatión Reductasa/química , Glutatión Reductasa/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/agonistas , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/química , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Sustancias Protectoras/metabolismo , Sustancias Protectoras/uso terapéutico , Ratas , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/química , Superóxido Dismutasa/metabolismo , Trehalosa/uso terapéutico
15.
J Med Chem ; 61(7): 2611-2635, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28876065

RESUMEN

Carbon monoxide (CO) is attracting increasing attention because of its role as a gasotransmitter with cytoprotective and homeostatic properties. Carbon monoxide releasing molecules (CORMs) are spatially and temporally controlled CO releasers that exhibit superior and more effective pharmaceutical traits than gaseous CO because of their chemistry and structure. Experimental and preclinical research in animal models has shown the therapeutic potential of inhaled CO and CORMs, and the biological effects of CO and CORMs have also been observed in preclinical trials via the genetic modulation of heme oxygenase-1 (HO-1). In this review, we describe the pharmaceutical use of CO and CORMs, methods of detecting CO release, and developments in CORM design and synthesis. Many valuable clinical CORMs formulated using macromolecules and nanomaterials are also described.


Asunto(s)
Antimetabolitos/química , Antimetabolitos/uso terapéutico , Monóxido de Carbono/química , Monóxido de Carbono/uso terapéutico , Animales , Antimetabolitos/administración & dosificación , Monóxido de Carbono/administración & dosificación , Complejos de Coordinación , Preparaciones de Acción Retardada , Hemo-Oxigenasa 1/química , Humanos
16.
PLoS One ; 12(11): e0188344, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29155886

RESUMEN

It has recently been shown that signal peptide peptidase (SPP) can catalyze the intramembrane cleavage of heme oxygenase-1 (HO-1) that leads to translocation of HO-1 into the cytosol and nucleus. While there is consensus that translocated HO-1 promotes tumor progression and drug resistance, the physiological signals leading to SPP-mediated intramembrane cleavage of HO-1 and the specificity of the process remain unclear. In this study, we used co-immunoprecipitation and confocal laser scanning microscopy to investigate the translocation mechanism of HO-1 and its regulation by SPP. We show that HO-1 and the closely related HO-2 isoenzyme bind to SPP under normoxic conditions. Under hypoxic conditions SPP mediates intramembrane cleavage of HO-1, but not HO-2. In experiments with an inactive HO-1 mutant (H25A) we show that translocation is independent of the catalytic activity of HO-1. Studies with HO-1 / HO-2 chimeras indicate that the membrane anchor, the PEST-domain and the nuclear shuttle sequence of HO-1 are necessary for full cleavage and subsequent translocation under hypoxic conditions. In the presence of co-expressed exogenous SPP, the anchor and the PEST-domain are sufficient for translocation. Taken together, we identified the domains involved in HO-1 translocation and showed that SPP-mediated cleavage is isoform-specific and independent of HO-activity. A closer understanding of the translocation mechanism of HO-1 is of particular importance because nuclear HO-1 seems to lead to tumor progression and drug resistance.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Membrana Celular/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas Mutantes Quiméricas/metabolismo , Secuencia de Aminoácidos , Ácido Aspártico Endopeptidasas/genética , Hipoxia de la Célula , Membrana Celular/química , Clonación Molecular , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Células HEK293 , Hemo Oxigenasa (Desciclizante)/química , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/genética , Humanos , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteolisis , Alineación de Secuencia , Homología de Secuencia de Aminoácido
17.
ACS Nano ; 11(12): 12641-12649, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29149552

RESUMEN

The discovery of biocompatible or bioactive nanoparticles for medicinal applications is an expensive and time-consuming process that may be significantly facilitated by incorporating more rational approaches combining both experimental and computational methods. However, it is currently hindered by two limitations: (1) the lack of high-quality comprehensive data for computational modeling and (2) the lack of an effective modeling method for the complex nanomaterial structures. In this study, we tackled both issues by first synthesizing a large library of nanoparticles and obtained comprehensive data on their characterizations and bioactivities. Meanwhile, we virtually simulated each individual nanoparticle in this library by calculating their nanostructural characteristics and built models that correlate their nanostructure diversity to the corresponding biological activities. The resulting models were then used to predict and design nanoparticles with desired bioactivities. The experimental testing results of the designed nanoparticles were consistent with the model predictions. These findings demonstrate that rational design approaches combining high-quality nanoparticle libraries, big experimental data sets, and intelligent computational models can significantly reduce the efforts and costs of nanomaterial discovery.


Asunto(s)
Materiales Biocompatibles/química , Oro/química , Modelos Químicos , Nanoestructuras/química , Línea Celular Tumoral , Células HEK293 , Hemo-Oxigenasa 1/química , Humanos , Relación Estructura-Actividad , Propiedades de Superficie
18.
PLoS One ; 12(7): e0181117, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28704474

RESUMEN

Heme is an efficient source of iron in the diet, and heme preparations are used to prevent and cure iron deficiency anemia in humans and animals. However, the molecular mechanisms responsible for heme absorption remain only partially characterized. Here, we employed young iron-deficient piglets as a convenient animal model to determine the efficacy of oral heme iron supplementation and investigate the pathways of heme iron absorption. The use of bovine hemoglobin as a dietary source of heme iron was found to efficiently counteract the development of iron deficiency anemia in piglets, although it did not fully rebalance their iron status. Our results revealed a concerted increase in the expression of genes responsible for apical and basolateral heme transport in the duodenum of piglets fed a heme-enriched diet. In these animals the catalytic activity of heme oxygenase 1 contributed to the release of elemental iron from the protoporphyrin ring of heme within enterocytes, which may then be transported by the strongly expressed ferroportin across the basolateral membrane to the circulation. We hypothesize that the well-recognized high bioavailability of heme iron may depend on a split pathway mediating the transport of heme-derived elemental iron and intact heme from the interior of duodenal enterocytes to the bloodstream.


Asunto(s)
Anemia Ferropénica/dietoterapia , Duodeno/metabolismo , Perfilación de la Expresión Génica/métodos , Hemo-Oxigenasa 1/genética , Hemo/administración & dosificación , Administración Oral , Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Hemo/uso terapéutico , Hemo-Oxigenasa 1/química , Humanos , Porcinos
19.
J Biol Chem ; 292(28): 11970-11979, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28588027

RESUMEN

An imbalance between oxidative stress and antioxidant activity plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Cigarette smoke, a major risk factor of COPD, induces cellular oxidative stress, but levels of antioxidants such as heme oxygenase-1 (HO-1) are reduced in individuals with severe COPD. In this study, we evaluated the molecular mechanism of reduced HO-1 expression in human bronchial epithelial cells. We found that cigarette smoke extract (CSE) increases HO-1 levels via activation of NFE2-related factor 2 (Nrf2). However, pretreating cells with the protease neutrophil elastase (NE) suppressed the CSE-induced expression of HO-1 mRNA and protein. NE also decreased the sirtuin 1 (SIRT1) level, but did not inhibit CSE-induced nuclear translocation and DNA-binding activity of Nrf2. Transfection of cells with a Myc/His-tagged SIRT1 expression vector completely blocked the NE-mediated suppression of HO-1 expression. We further noted that the NE-induced down-regulation of SIRT1 was not due to decreased transcription or proteasomal/lysosomal degradation or loss of solubility. Immunofluorescence staining revealed that NE enters the cell cytoplasm, and we observed that NE directly cleaved SIRT1 in vitro, indicating that SIRT1 levels are decreased via direct degradation by internalized NE. Of note, we observed decreased SIRT1 levels in NE-treated primary human bronchial epithelial cells and in lung homogenates from both smokers and patients with COPD. In conclusion, NE suppresses CSE-induced HO-1 expression by cleaving SIRT1. This finding indicates the importance of cross-talk between oxidative stress and protease responses in the pathogenesis of COPD.


Asunto(s)
Bronquios/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Elastasa de Leucocito/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Sirtuina 1/metabolismo , Fumar/efectos adversos , Transporte Activo de Núcleo Celular , Biomarcadores/metabolismo , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/patología , Línea Celular , Células Cultivadas , Mezclas Complejas/toxicidad , Regulación Enzimológica de la Expresión Génica , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/química , Humanos , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Estrés Oxidativo , Transporte de Proteínas , Proteolisis , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Proteínas Recombinantes de Fusión , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/química , Sirtuina 1/genética , Humo/efectos adversos , Humo/análisis , Fumar/metabolismo , Fumar/patología , Productos de Tabaco/efectos adversos , Productos de Tabaco/análisis
20.
J Biol Chem ; 292(32): 13205-13229, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28655775

RESUMEN

Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Moreover, intestinal iron is incorporated as ferrous iron, which is transported via the iron importer, divalent metal transporter 1 (DMT1). Recently, we demonstrated that the iron chaperone poly(rC)-binding protein 2 (PCBP2) can directly receive ferrous iron from DMT1 or transfer iron to the iron exporter, ferroportin 1. To promote intracellular iron flux, an iron chaperone may be essential for receiving iron generated by heme catabolism, but this hypothesis is untested so far. Herein, we demonstrate that HO1 binds to PCBP2, but not to other PCBP family members, namely PCBP1, PCBP3, or PCBP4. Interestingly, HO1 formed a complex with either CPR or PCBP2, and it was demonstrated that PCBP2 competes with CPR for HO1 binding. Using PCBP2-deletion mutants, we demonstrated that the PCBP2 K homology 3 domain is important for the HO1/PCBP2 interaction. In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Furthermore, in vitro reconstitution experiments with purified recombinant proteins indicated that HO1 could bind to PCBP2 in the presence of heme, whereas loading of PCBP2 with ferrous iron caused PCBP2 to lose its affinity for HO1. These results indicate that ferrous iron released from heme can be bound by PCBP2 and suggest a model for an integrated heme catabolism and iron transport metabolon.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hemo/metabolismo , Hierro/metabolismo , NADPH-Ferrihemoproteína Reductasa/metabolismo , Proteínas de Unión al ARN/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Unión Competitiva , Transporte Biológico , Línea Celular , Eliminación de Gen , Hemo Oxigenasa (Desciclizante)/química , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/genética , Humanos , Metaloporfirinas/metabolismo , Mutación , NADPH-Ferrihemoproteína Reductasa/antagonistas & inhibidores , NADPH-Ferrihemoproteína Reductasa/química , NADPH-Ferrihemoproteína Reductasa/genética , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Transporte de Proteínas , Interferencia de ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Homología Estructural de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...