Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(8): 105009, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37406814

RESUMEN

Selenoprotein P (SeP, encoded by the SELENOP gene) is a plasma protein that contains selenium in the form of selenocysteine residues (Sec, a cysteine analog containing selenium instead of sulfur). SeP functions for the transport of selenium to specific tissues in a receptor-dependent manner. Apolipoprotein E receptor 2 (ApoER2) has been identified as a SeP receptor. However, diverse variants of ApoER2 have been reported, and the details of its tissue specificity and the molecular mechanism of its efficiency remain unclear. In the present study, we found that human T lymphoma Jurkat cells have a high ability to utilize selenium via SeP, while this ability was low in human rhabdomyosarcoma cells. We identified an ApoER2 variant with a high affinity for SeP in Jurkat cells. This variant had a dissociation constant value of 0.67 nM and a highly glycosylated O-linked sugar domain. Moreover, the acidification of intracellular vesicles was necessary for selenium transport via SeP in both cell types. In rhabdomyosarcoma cells, SeP underwent proteolytic degradation in lysosomes and transported selenium in a Sec lyase-dependent manner. However, in Jurkat cells, SeP transported selenium in Sec lyase-independent manner. These findings indicate a preferential selenium transport pathway involving SeP and high-affinity ApoER2 in a Sec lyase-independent manner. Herein, we provide a novel dynamic transport pathway for selenium via SeP.


Asunto(s)
Liasas , Selenio , Humanos , Liasas/metabolismo , Selenio/metabolismo , Selenocisteína/genética , Selenocisteína/metabolismo , Selenoproteína P/genética , Selenoproteína P/metabolismo , Selenoproteínas , Células Jurkat
2.
Cell Struct Funct ; 48(1): 59-70, 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36575042

RESUMEN

Stimulator of interferon genes (STING) is an ER-localized transmembrane protein and the receptor for 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which is a second messenger produced by cGAMP synthase (cGAS), a cytosolic double-stranded DNA sensor. The cGAS-STING pathway plays a critical role in the innate immune response to infection of a variety of DNA pathogens through the induction of the type I interferons. Pharmacological activation of STING is a promising therapeutic strategy for cancer, thus the development of potent and selective STING agonists has been pursued. Here we report that mouse STING can be activated by phenylarsine oxide (PAO), a membrane permeable trivalent arsenic compound that preferentially reacts with thiol group of cysteine residue (Cys). The activation of STING with PAO does not require cGAS or cGAMP. Mass spectrometric analysis of the peptides generated by trypsin and chymotrypsin digestion of STING identifies several PAO adducts, suggesting that PAO covalently binds to STING. Screening of STING variants with single Cys to serine residues (Ser) reveals that Cys88 and Cys291 are critical to the response to PAO. STING activation with PAO, as with cGAMP, requires the ER-to-Golgi traffic and palmitoylation of STING. Our results identify a non-nucleotide STING agonist that does not target the cGAMP-binding pocket, and demonstrate that Cys of STING can be a novel target for the development of STING agonist.Key words: STING agonist, cysteine modification, innate immunity, phenylarsine oxide.


Asunto(s)
Cisteína , Transducción de Señal , Ratones , Animales , Proteínas de la Membrana/metabolismo , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , ADN
3.
Biol Pharm Bull ; 46(9): 1162-1168, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37661394

RESUMEN

Methylmercury (MeHg) is the causal substrate of Minamata disease and a major environmental toxicant. MeHg is widely distributed, mainly in the ocean, meaning its bioaccumulation in seafood is a considerable problem for human health. MeHg has been intensively investigated and is known to induce inflammatory responses and neurodegeneration. However, the relationship between MeHg-induced inflammatory responses and neurodegeneration is not understood. In the present review, we first describe recent findings showing an association between inflammatory responses and certain MeHg-unrelated neurological diseases caused by neurodegeneration. In addition, cell-specific MeHg-induced inflammatory responses are summarized for the central nervous system including those of microglia, astrocytes, and neurons. We also describe MeHg-induced inflammatory responses in peripheral cells and tissue, such as macrophages and blood. These findings provide a concept of the relationship between MeHg-induced inflammatory responses and neurodegeneration, as well as direction for future research of MeHg-induced neurotoxicity.


Asunto(s)
Compuestos de Metilmercurio , Síndromes de Neurotoxicidad , Humanos , Compuestos de Metilmercurio/toxicidad , Síndromes de Neurotoxicidad/etiología , Inflamación/inducido químicamente , Astrocitos , Sistema Nervioso Central
4.
Arch Toxicol ; 97(7): 1887-1897, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37193757

RESUMEN

We previously found that methylmercury induces expression of oncostatin M (OSM), which is released extracellularly and binds to tumor necrosis factor receptor 3 (TNFR3), possibly enhancing its own toxicity. However, the mechanism by which methylmercury causes OSM to bind to TNFR3 rather than to its known receptors, OSM receptor and LIFR, is unknown. In this study, we aimed to elucidate the effect of methylmercury modification of cysteine residues in OSM on binding to TNFR3. Immunostaining of TNFR3-V5-expressing cells suggested that methylmercury promoted binding of OSM to TNFR3 on the cell membrane. In an in vitro binding assay, OSM directly bound to the extracellular domain of TNFR3, and this binding was promoted by methylmercury. Additionally, the formation of a disulfide bond in the OSM molecule was essential for the binding of both proteins, and LC/MS analysis revealed that methylmercury directly modified the 105th cysteine residue (Cys105) in OSM. Next, mutant OSM, in which Cys105 was replaced by serine or methionine, increased the binding to TNFR3, and a similar effect was observed in immunoprecipitation using cultured cells. Furthermore, cell proliferation was inhibited by treatment with Cys105 mutant OSMs compared with wildtype OSM, and this effect was cancelled by TNFR3 knockdown. In conclusion, we revealed a novel mechanism of methylmercury toxicity, in which methylmercury directly modifies Cys105 in OSM, thereby inhibiting cell proliferation via promoting binding to TNFR3. This indicates a chemical disruption in the interaction between the ligand and the receptor is a part of methylmercury toxicity.


Asunto(s)
Cisteína , Compuestos de Metilmercurio , Oncostatina M/química , Oncostatina M/metabolismo , Compuestos de Metilmercurio/toxicidad , Receptores del Factor de Necrosis Tumoral , Proliferación Celular
5.
Nucleic Acids Res ; 49(12): 6893-6907, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34142161

RESUMEN

Selenoprotein P (SELENOP) is a major plasma selenoprotein that contains 10 Sec residues, which is encoded by the UGA stop codon. The mRNA for SELENOP has the unique property of containing two Sec insertion sequence (SECIS) elements, which is located in the 3' untranslated region (3'UTR). Here, we coincidentally identified a novel gene, CCDC152, by sequence analysis. This gene was located in the antisense region of the SELENOP gene, including the 3'UTR region in the genome. We demonstrated that this novel gene functioned as a long non-coding RNA (lncRNA) that decreased SELENOP protein levels via translational rather than transcriptional, regulation. We found that the CCDC152 RNA interacted specifically and directly with the SELENOP mRNA and inhibited its binding to the SECIS-binding protein 2, resulting in the decrease of ribosome binding. We termed this novel gene product lncRNA inhibitor of SELENOP translation (L-IST). Finally, we found that epigallocatechin gallate upregulated L-IST in vitro and in vivo, to suppress SELENOP protein levels. Here, we provide a new regulatory mechanism of SELENOP translation by an endogenous long antisense ncRNA.


Asunto(s)
Regulación de la Expresión Génica , Biosíntesis de Proteínas , ARN Largo no Codificante/metabolismo , Selenoproteína P/genética , Catequina/análogos & derivados , Catequina/farmacología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , ARN Largo no Codificante/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Selenoproteína P/biosíntesis
6.
Biochem J ; 478(7): 1453-1470, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33749780

RESUMEN

Redox regulation of proteins via cysteine residue oxidation is involved in the control of various cellular signal pathways. Pyruvate kinase M2 (PKM2), a rate-limiting enzyme in glycolysis, is critical for the metabolic shift from glycolysis to the pentose phosphate pathway under oxidative stress in cancer cell growth. The PKM2 tetramer is required for optimal pyruvate kinase (PK) activity, whereas the inhibition of inter-subunit interaction of PKM2 induced by Cys358 oxidation has reduced PK activity. In the present study, we identified three oxidation-sensitive cysteine residues (Cys358, Cys423 and Cys424) responsible for four oxidation forms via the thiol oxidant diamide and/or hydrogen peroxide (H2O2). Possibly due to obstruction of the dimer-dimer interface, H2O2-induced sulfenylation (-SOH) and diamide-induced modification at Cys424 inhibited tetramer formation and PK activity. Cys423 is responsible for intermolecular disulfide bonds with heterologous proteins via diamide. Additionally, intramolecular polysulphide linkage (-Sn-, n ≧ 3) between Cys358 and an unidentified PKM2 Cys could be induced by diamide. We observed that cells expressing the oxidation-resistant PKM2 (PKM2C358,424A) produced more intracellular reactive oxygen species (ROS) and exhibited greater sensitivity to ROS-generating reagents and ROS-inducible anti-cancer drugs compared with cells expressing wild-type PKM2. These results highlight the possibility that PKM2 inhibition via Cys358 and Cys424 oxidation contributes to eliminating excess ROS and oxidative stress.


Asunto(s)
Proteínas Portadoras/química , Cisteína/química , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/patología , Proteínas de la Membrana/química , Estrés Oxidativo , Compuestos de Sulfhidrilo/química , Hormonas Tiroideas/química , Proteínas Portadoras/metabolismo , Glucólisis , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Hormonas Tiroideas/metabolismo , Células Tumorales Cultivadas , Proteínas de Unión a Hormona Tiroide
7.
Biol Pharm Bull ; 43(10): 1562-1569, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32999166

RESUMEN

trans-Fatty acids (TFAs) are unsaturated fatty acids with at least one carbon-carbon double bond in trans configuration. TFA consumption has been epidemiologically associated with neurodegenerative diseases (NDs) including Alzheimer's disease. However, the underlying mechanisms of TFA-related NDs remain unknown. Here, we show a novel microglial signaling pathway that induces inflammation and cell death, which is dramatically enhanced by elaidic acid (EA), the most abundant TFA derived from food. We found that extracellular ATP, one of the damage-associated molecular patterns (DAMPs) leaked from injured cells, induced activation of the apoptosis signal-regulating kinase 1 (ASK1)-p38 pathway, which is one of the major stress-responsive mitogen-activated protein (MAP) kinase signaling pathways, and subsequent caspase-3 cleavage and DNA ladder formation (hallmarks of apoptosis) in mouse microglial cell lines including BV2 and MG6 cells. Furthermore, we found that in these microglial cell lines, EA, but not its cis isomer oleic acid, facilitated extracellular ATP-induced ASK1/p38 activation and apoptosis, which was suppressed by pharmacological inhibition of either p38, reactive oxygen species (ROS) generation, P2X purinoceptor 7 (P2X7), or Ca2+/calmodulin-dependent kinase II (CaMKII). These results demonstrate that in microglial cells, extracellular ATP induces activation of the ASK1-p38 MAP kinase pathway and ultimately apoptosis downstream of P2X7 receptor and ROS generation, and that EA promotes ATP-induced apoptosis through CaMKII-dependent hyperactivation of the ASK1-p38 pathway, in the same manner as in macrophages. Our study may provide an insight into the pathogenesis of NDs associated with TFAs.


Asunto(s)
Adenosina Trifosfato/administración & dosificación , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Ácidos Oléicos/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Microglía/metabolismo
8.
Toxicol Appl Pharmacol ; 356: 120-126, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30081057

RESUMEN

We previously reported significantly increased level of putrescine, a polyamine, in the brains of mice administered methylmercury. Moreover, addition of putrescine to culture medium reduced methylmercury toxicity in C17.2 mouse neural stem cells. In this study, the role of ornithine decarboxylase (ODC), an enzyme involved in putrescine synthesis, in response to methylmercury toxicity was investigated. Methylmercury increased ODC activity in mouse cerebrum and cerebellum, but this increase was hardly observed in the kidney and liver, where methylmercury accumulated at a high concentration. In the cerebrum and cerebellum, increased putrescine was observed with methylmercury administration. Methylmercury increased ODC activity in C17.2 cells, but this was almost completely abolished in the presence of an ODC inhibitor. Methylmercury also increased the level of ODC protein in mouse brain and C17.2 cells. In addition, C17.2 cells pretreated with ODC inhibitor showed higher methylmercury sensitivity than control cells. These results suggest that the increased ODC activity by methylmercury is involved in the increase in putrescine level, and ODC plays an important role in the reduction of methylmercury toxicity. This is the first study to provide evidence that increased ODC activity may be a protective response against methylmercury-induced neurotoxicity.


Asunto(s)
Activación Enzimática/efectos de los fármacos , Intoxicación por Mercurio/metabolismo , Intoxicación por Mercurio/prevención & control , Compuestos de Metilmercurio/toxicidad , Ornitina Descarboxilasa/efectos de los fármacos , Putrescina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Línea Celular , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Mercurio/farmacocinética , Ratones , Células-Madre Neurales , Inhibidores de la Ornitina Descarboxilasa/farmacología , Distribución Tisular
9.
Chem Res Toxicol ; 30(9): 1673-1684, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28837763

RESUMEN

Electrophiles such as methylmercury (MeHg) affect cellular functions by covalent modification with endogenous thiols. Reactive persulfide species were recently reported to mediate antioxidant responses and redox signaling because of their strong nucleophilicity. In this study, we used MeHg as an environmental electrophile and found that exposure of cells to the exogenous electrophile elevated intracellular concentrations of the endogenous electrophilic molecule 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), accompanied by depletion of reactive persulfide species and 8-SH-cGMP which is a metabolite of 8-nitro-cGMP. Exposure to MeHg also induced S-guanylation and activation of H-Ras followed by injury to cerebellar granule neurons. The electrophile-induced activation of redox signaling and the consequent cell damage were attenuated by pretreatment with a reactive persulfide species donor. In conclusion, exogenous electrophiles such as MeHg with strong electrophilicity impair the redox signaling regulatory mechanism, particularly of intracellular reactive persulfide species and therefore lead to cellular pathogenesis. Our results suggest that reactive persulfide species may be potential therapeutic targets for attenuating cell injury by electrophiles.


Asunto(s)
Compuestos de Metilmercurio/química , Sulfuros/química , Animales , Anticuerpos/inmunología , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , GMP Cíclico/análogos & derivados , GMP Cíclico/química , GMP Cíclico/inmunología , GMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inmunohistoquímica , Masculino , Compuestos de Metilmercurio/análisis , Compuestos de Metilmercurio/toxicidad , Microscopía Fluorescente , Naftoquinonas/química , Naftoquinonas/toxicidad , Óxido Nítrico/análisis , Oxidación-Reducción , Células PC12 , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Espectrometría de Masa por Ionización de Electrospray , Sulfuros/farmacología , Proteínas ras/genética , Proteínas ras/metabolismo
10.
J Mol Cell Cardiol ; 73: 10-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24530900

RESUMEN

Structural and morphological changes of the cardiovascular systems (cardiovascular remodeling) are a major clinical outcome of cardiovascular diseases. Many lines of evidences have implied that transfiguration of reduction/oxidation (redox) homeostasis due to excess production of reactive oxygen species (ROS) and/or ROS-derived electrophilic metabolites (electrophiles) is the main cause of cardiovascular remodeling. Gasotransmitters, such as nitric oxide (NO) and endogenous electrophiles, are considered major bioactive species and have been extensively studied in the context of physiological and pathological cardiovascular events. We have recently found that hydrogen sulfide-related reactive species function as potent nucleophiles to eliminate electrophilic modification of signaling proteins induced by NO-derived electrophilic byproducts (e.g., 8-nitroguanosine 3',5'-cyclic monophosphate and nitro-oleic acid). In this review, we discuss the current understanding of redox control of cardiovascular pathophysiology by electrophiles and nucleophiles. We propose that modulation of electrophile-mediated post-translational modification of protein cysteine thiols may be a new therapeutic strategy of cardiovascular diseases. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".


Asunto(s)
GMP Cíclico/análogos & derivados , Animales , GMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
11.
J Neurochem ; 130(6): 826-38, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24832624

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disease, but its pathogenesis remains elusive. A mutation in ubiquitin C-terminal hydrolase L1 (UCH-L1) is responsible for a form of genetic PD which strongly resembles the idiopathic PD. We previously showed that 1-(3',4'-dihydroxybenzyl)-1,2,3,4-tetrahydroisoquinoline (3',4'DHBnTIQ) is an endogenous parkinsonism-inducing dopamine derivative. Here, we investigated the interaction between 3',4'DHBnTIQ and UCH-L1 and its possible role in the pathogenesis of idiopathic PD. Our results indicate that 3',4'DHBnTIQ binds to UCH-L1 specifically at Cys152 in vitro. In addition, 3',4'DHBnTIQ treatment increased the amount of UCH-L1 in the insoluble fraction of SH-SY5Y cells and inhibited its hydrolase activity to 60%, reducing the level of ubiquitin in the soluble fraction of SH-SY5Y cells. Catechol-modified UCH-L1 as well as insoluble UCH-L1 were detected in the midbrain of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated PD model mice. Structurally as well as functionally altered UCH-L1 have been detected in the brains of patients with idiopathic PD. We suggest that conjugation of UCH-L1 by neurotoxic endogenous compounds such as 3',4'DHBnTIQ might play a key role in onset and progression of idiopathic PD. We investigated the interaction between ubiquitin C-terminal hydrolase L1 (UCH-L1) and the brain endogenous parkinsonism inducer 1-(3',4'-dihydroxybenzyl)-1,2,3,4-tetrahydroisoquinoline (3',4'DHBnTIQ). Our results indicate that 3',4'DHBnTIQ binds to UCH-L1 specifically at cysteine 152 and induces its aggregation. 3',4'DHBnTIQ also inhibits the hydrolase activity of UCH-L1. Catechol-modified as well as insoluble UCH-L1 were detected in the midbrains of MPTP-treated Parkinson's disease (PD) model mice. Conjugation of UCH-L1 by neurotoxic endogenous compounds like 3',4'DHBnTIQ might play a key role in onset and progression of PD.


Asunto(s)
Dopamina/análogos & derivados , Dopamina/metabolismo , Neurotoxinas/metabolismo , Enfermedad de Parkinson/metabolismo , Tretoquinol/análogos & derivados , Ubiquitina Tiolesterasa/metabolismo , Animales , Western Blotting , Catecoles/química , Catecoles/farmacología , Línea Celular Tumoral , Supervivencia Celular , Electroforesis en Gel de Agar , Escherichia coli/metabolismo , Humanos , Indicadores y Reactivos , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tretoquinol/metabolismo , Tretoquinol/farmacología , Ubiquitina Tiolesterasa/química
12.
Chem Res Toxicol ; 27(1): 76-85, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24443938

RESUMEN

Polycyclic aromatic hydrocarbon quinones (PAHQs) are components in airborne particulate matter (PM) and generate reactive oxygen species (ROS) in a redox cycling process. 9,10-Phenanthrenequinone (9,10-PQ) is a PAHQ found in diesel exhaust particulates and PM. When inhaled, it produces much more ROS than other PAHQs. We hypothesized that urinary metabolites of 9,10-PQ could serve as biomarkers of PAHQ exposure. Here, we describe methods for pretreating urine samples and analyzing 9,10-PQ metabolites by liquid chromatography with tandem mass spectrometry (LC-MS/MS). In urine from rats intraperitoneally injected with 9,10-PQ, the monoglucuronide of 9,10-dihydroxyphenanthrene (9,10-PQHG) was found to be a major metabolite of 9,10-PQ. 9,10-PQHG was also identified in the urine of a nonoccupationally exposed human by its retention time and MS/MS spectra. Furthermore, the urine contained hardly any free (unmetabolized) 9,10-PQ, but treating it with hydrolytic enzymes released 9,10-PQ from conjugated metabolites such as 9,10-PQHG. The concentrations of 9,10-PQHG in urine samples from nonoccupationally exposed subjects who lived in a suburban area were 2.04-19.08 nmol/mol creatinine. This study is the first to demonstrate the presence of 9,10-PQHG in human urine. Determination of urinary 9,10-PQHG should be useful for determining 9,10-PQ exposure.


Asunto(s)
Fenantrenos/metabolismo , Fenantrenos/orina , Especies Reactivas de Oxígeno/metabolismo , Adulto , Animales , Cromatografía Liquida , Femenino , Humanos , Masculino , Estructura Molecular , Fenantrenos/química , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Adulto Joven
13.
Hepatol Res ; 44(10): E84-91, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23957840

RESUMEN

AIM: Anemia frequently develops in patients given pegylated interferon, ribavirin (RBV), telaprevir (TVR) triple therapy and restricts treatment by forcing reduction or discontinuation of RBV administration. We investigated whether erythropoietin (EPO) could alleviate RBV-induced anemia to help maintain the RBV dose during the first 12 weeks, the triple therapy phase. METHODS: Twenty-two patients with hepatitis C virus (HCV) genotype 1 were enrolled. Hemoglobin (Hb) concentration was measured every week. If Hb reduction from the baseline was 2 g/dL or more, 12 000 IU of epoetin-α was administrated. When further reduction (≥3 g/dL) was observed, 24 000 IU of epoetin-α was used. Inosine triphosphatase (ITPA) single nucleotide polymorphism (rs1127354) was genotyped for all patients. RESULTS: Among the 22 patients enrolled in this study, three required RBV dose reduction due to anemia, two had to discontinue or reduce TVR and RBV due to creatinine elevation. The remaining 17 patients completed the treatment during the triple therapy phase without reduction of the RBV dose or adverse events attributable to EPO. Regardless of ITPA genotype, Hb decline was well controlled by EPO administration, whereas the total EPO dose tended to be higher in the CC genotype group. The average adherence to RBV during the triple therapy phase was 97.5%. SVR was achieved in 17 patients; two patients had viral breakthrough and three patients had relapse of HCV RNA. CONCLUSION: EPO can be a favorable alternative to reduction of RBV to facilitate the adherence of patients on TVR-based triple therapy.

14.
Yakugaku Zasshi ; 144(1): 41-45, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38171793

RESUMEN

Methylmercury is a ubiquitous neurotoxic substance present in the environment, and health concerns, especially through the consumption of seafood, remain. Glutathione (GSH)-mediated detoxification and the excretion of methylmercury are known metabolic detoxification pathways. We have also discovered a mechanism by which endogenous super-sulfides convert methylmercury to nontoxic metabolites such as bis-methylmercury sulfide. However, these metabolites are present in very small quantities, and the significance of the detoxification of methylmercury by super-sulfides is not well understood. Methylmercury binds to thiol groups in vivo but can also react with highly reactive selenols (selenocysteine residues). Such covalent bonds (S-mercuration and Se-mercuration) are broken by nucleophilic substitution reactions with other thiol and selenols, however, the contribution of super-sulfides to this substitution reaction is not well understood. Interestingly, a recent study suggested that selenoprotein P, the major selenium transport protein in plasma, binds to methylmercury, however, Se-mercuration was not determined. In this review, we introduce these series of reactions and discuss their involvement with super-sulfides in methylmercury toxicity.


Asunto(s)
Compuestos de Metilmercurio , Selenio , Compuestos de Metilmercurio/metabolismo , Selenio/metabolismo , Glutatión/metabolismo , Compuestos de Sulfhidrilo , Sulfuros
15.
Sci Rep ; 14(1): 682, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38182643

RESUMEN

Glioblastoma (GBM) is one of the most aggressive and deadly brain tumors; however, its current therapeutic strategies are limited. Selenoprotein P (SeP; SELENOP, encoded by the SELENOP gene) is a unique selenium-containing protein that exhibits high expression levels in astroglia. SeP is thought to be associated with ferroptosis sensitivity through the induction of glutathione peroxidase 4 (GPX4) via selenium supplementation. In this study, to elucidate the role of SeP in GBM, we analyzed its expression in GBM patients and found that SeP expression levels were significantly higher when compared to healthy subjects. Knock down of SeP in cultured GBM cells resulted in a decrease in GPX1 and GPX4 protein levels. Under the same conditions, cell death caused by RSL3, a ferroptosis inducer, was enhanced, however this enhancement was canceled by supplementation of selenite. These results indicate that SeP expression contributes to preserving GPX and selenium levels in an autocrine/paracrine manner, i.e., SeP regulates a dynamic cycling-selenium storage system in GBM. We also confirmed the role of SeP expression in ferroptosis sensitivity using patient-derived primary GBM cells. These findings indicate that expression of SeP in GBM can be a significant therapeutic target to overcome anticancer drug resistance.


Asunto(s)
Ferroptosis , Glioblastoma , Selenio , Selenoproteína P , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Selenio/metabolismo , Selenoproteína P/metabolismo
16.
Commun Biol ; 6(1): 1060, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37857700

RESUMEN

Selenoprotein P (SeP) is a major selenoprotein in serum predominantly produced in the liver. Excess SeP impairs insulin secretion from the pancreas and insulin sensitivity in skeletal muscle, thus inhibition of SeP could be a therapeutic strategy for type 2 diabetes. In this study, we examine the effect of sulforaphane (SFN), a phytochemical of broccoli sprouts and an Nrf2 activator, on SeP expression in vitro and in vivo. Treatment of HepG2 cells with SFN decreases inter- and intra-cellular SeP levels. SFN enhances lysosomal acidification and expression of V-ATPase, and inhibition of this process cancels the decrease of SeP by SFN. SFN activates Nrf2 in the cells, while Nrf2 siRNA does not affect the decrease of SeP by SFN or lysosomal acidification. These results indicate that SFN decreases SeP by enhancing lysosomal degradation, independent of Nrf2. Injection of SFN to mice results in induction of cathepsin and a decrease of SeP in serum. The findings from this study are expected to contribute to developing SeP inhibitors in the future, thereby contributing to treating and preventing diseases related to increased SeP.


Asunto(s)
Diabetes Mellitus Tipo 2 , Factor 2 Relacionado con NF-E2 , Ratones , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Selenoproteína P , Lisosomas/metabolismo
17.
Arch Toxicol ; 86(11): 1693-702, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22752181

RESUMEN

We previously developed a screening method to identify proteins that undergo aggregation through S-mercuration by methylmercury (MeHg) and found that rat arginase I is a target protein for MeHg (Kanda et al. in Arch Toxicol 82:803-808, 2008). In the present study, we characterized another S-mercurated protein from a rat hepatic preparation that has a subunit mass of 42 kDa, thereby facilitating its aggregation. Two-dimensional SDS-polyacrylamide gel electrophoresis and subsequent peptide mass fingerprinting using matrix-assisted laser desorption and ionization time-of-flight mass spectrometry revealed that the 42 kDa protein was NAD-dependent sorbitol dehydrogenase (SDH). With recombinant rat SDH, we found that MeHg is covalently bound to SDH through Cys44, Cys119, Cys129 and Cys164, resulting in the inhibition of its catalytic activity, release of zinc ions and facilitates protein aggregation. Mutation analysis indicated that Cys44, which ligates the active site zinc atom, and Cys129 play a crucial role in the MeHg-mediated aggregation of SDH. Pretreatment with the cofactor NAD, but not NADP or FAD, markedly prevented aggregation of SDH. Such a protective effect of NAD on the aggregation of SDH caused by MeHg is discussed.


Asunto(s)
L-Iditol 2-Deshidrogenasa/química , L-Iditol 2-Deshidrogenasa/metabolismo , Compuestos de Metilmercurio/química , Zinc/metabolismo , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Cisteína/química , Cisteína/genética , Electroforesis en Gel Bidimensional , L-Iditol 2-Deshidrogenasa/genética , Hígado/enzimología , Datos de Secuencia Molecular , Mutación , NAD/metabolismo , Mapeo Peptídico , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Zinc/química
18.
Free Radic Biol Med ; 183: 89-103, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35318102

RESUMEN

Selenoprotein P (SELENOP) is a major selenium (Se)-containing protein (selenoprotein) in human plasma that is mainly synthesized in the liver. SELENOP transports Se to the cells, while SELENOP synthesized in peripheral tissues is incorporated in a paracrine/autocrine manner to maintain the levels of cellular selenoproteins, called the SELENOP cycle. Pancreatic ß cells, responsible for the synthesis and secretion of insulin, are known to express SELENOP. Here, using MIN6 cells as a mouse model for pancreatic ß cells and Selenop small interfering (si)RNA, we found that Selenop gene knockdown (KD) resulted in decreased cell viability, cellular pro/insulin levels, insulin secretion, and levels of several cellular selenoproteins, including glutathione peroxidase 4 (Gpx4) and selenoprotein K (Selenok). These dysfunctions induced by Selenop siRNA were recovered by the addition of Se. Ferroptosis-like cell death, regulated by Gpx4, was involved in the decrease of cell viability by Selenop KD, while stress-induced nascent granule degradation (SINGD), regulated by Selenok, was responsible for the decrease in proinsulin. SINGD was also observed in the pancreatic ß cells of Selenop knockout mice. These findings indicate a significant role of SELENOP expression for the function of pancreatic ß cells by maintaining the levels of cellular selenoproteins such as GPX4 and SELENOK.


Asunto(s)
Ferroptosis , Células Secretoras de Insulina , Selenio , Selenoproteína P , Animales , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Selenio/metabolismo , Selenoproteína P/genética , Selenoproteína P/metabolismo
19.
Chem Res Toxicol ; 24(10): 1633-5, 2011 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-21951228

RESUMEN

Methylmercury (MeHg) covalently modifies cellular proteins through their SH groups, resulting in cytotoxicity. We report that cystathionine ß-synthase (CBS), which catalyzes the production of hydrogen sulfide, contributes to cellular protection against MeHg. Pretreatment with NaHS or overexpression of CBS reduced MeHg cytotoxicity, whereas transfection with CBS small interfering RNA enhanced MeHg toxicity in human neuroblastoma SH-SY5Y cells. Bismethylmercury sulfide ((MeHg)(2)S) was identified as a metabolite of MeHg in SH-SY5Y cells exposed to MeHg and in the livers of rats treated with MeHg. (MeHg)(2)S had little chemical protein modification capability and little cytotoxicity compared with MeHg in vitro and in vivo.


Asunto(s)
Cistationina betasintasa/metabolismo , Contaminantes Ambientales/toxicidad , Sulfuro de Hidrógeno/metabolismo , Compuestos de Metilmercurio/toxicidad , Animales , Línea Celular Tumoral , Contaminantes Ambientales/metabolismo , Contaminantes Ambientales/farmacocinética , Humanos , Inactivación Metabólica , Hígado/metabolismo , Compuestos de Metilmercurio/metabolismo , Compuestos de Metilmercurio/farmacocinética , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Sustancias Protectoras/farmacología , Ratas , Sulfuros/farmacología
20.
Gan To Kagaku Ryoho ; 38(11): 1857-9, 2011 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-22083197

RESUMEN

A 66-year-old man was referred to our hospital with obstructive jaundice. Computed tomography(CT)scan showed thickening of the gallbladder wall, invasion into the liver bed, and thickening of the rectal wall. Colonoscopy revealed a type 2 rectal cancer, in which adenocarcinoma was identified by endoscopic biopsy. He was diagnosed with double-cancer of the gallbladder and rectum. Because his gallbladder cancer was more life threatening than his rectal cancer, gemcitabine was administered at 1, 000 mg/m2 on days 1, 8, and 15 of a 28-day course. After 3 courses of gemcitabine, the CT scan showed that the lymph nodes in the hepatoduodenal ligament had been enlarged, and duodenal stenosis had occurred as a result of gallbladder cancer invasion. S-1 was administered orally at doses of 120 mg/day twice daily on days 1-28 of a 42-day course. Partial response was confirmed by CT scan. After 8 courses of S-1, the gallbladder cancer had progressed and liver metastases had appeared. He subsequently died of disease progression. He survived for 17 months after the first course of chemotherapy, and the progression-free survival with S-1 was 10 months. Therefore, S-1 could be an effective agent for synchronous double cancer of the gallbladder and rectum.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias Primarias Múltiples/tratamiento farmacológico , Ácido Oxónico/uso terapéutico , Neoplasias del Recto/tratamiento farmacológico , Terapia Recuperativa , Tegafur/uso terapéutico , Anciano , Terapia Combinada , Combinación de Medicamentos , Resultado Fatal , Neoplasias de la Vesícula Biliar/patología , Neoplasias de la Vesícula Biliar/cirugía , Humanos , Masculino , Neoplasias Primarias Múltiples/patología , Neoplasias Primarias Múltiples/cirugía , Neoplasias del Recto/patología , Neoplasias del Recto/cirugía , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA