Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35217600

RESUMEN

An ideal cancer therapeutic strategy involves the selective killing of cancer cells without affecting the surrounding normal cells. However, researchers have failed to develop such methods for achieving selective cancer cell death because of shared features between cancerous and normal cells. In this study, we have developed a therapeutic strategy called the cancer-specific insertions-deletions (InDels) attacker (CINDELA) to selectively induce cancer cell death using the CRISPR-Cas system. CINDELA utilizes a previously unexplored idea of introducing CRISPR-mediated DNA double-strand breaks (DSBs) in a cancer-specific fashion to facilitate specific cell death. In particular, CINDELA targets multiple InDels with CRISPR-Cas9 to produce many DNA DSBs that result in cancer-specific cell death. As a proof of concept, we demonstrate here that CINDELA selectively kills human cancer cell lines, xenograft human tumors in mice, patient-derived glioblastoma, and lung patient-driven xenograft tumors without affecting healthy human cells or altering mouse growth.


Asunto(s)
Sistemas CRISPR-Cas , Mutación INDEL , Neoplasias/genética , Animales , Muerte Celular/genética , Roturas del ADN de Doble Cadena , Xenoinjertos , Humanos , Ratones
2.
J Cell Mol Med ; 22(1): 57-66, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782172

RESUMEN

3'-Sialyllactose has specific physiological functions in a variety of tissues; however, its effects on osteoarthritic development remain unknown. Here, we demonstrated the function of 3'-sialyllactose on osteoarthritic cartilage destruction. In vitro and ex vivo, biochemical and histological analysis demonstrated that 3'-sialyllactose was sufficient to restore the synthesis of Col2a1 and accumulation of sulphated proteoglycan, a critical factor for cartilage regeneration in osteoarthritic development, and blocked the expression of Mmp3, Mmp13 and Cox2 induced by IL-1ß, IL-6, IL-17 and TNF-α, which mediates cartilage degradation. Further, reporter gene assays revealed that the activity of Sox9 as a transcription factor for Col2a1 expression was accelerated by 3'-sialyllactose, whereas the direct binding of NF-κB to the Mmp3, Mmp13 and Cox2 promoters was reduced by 3'-sialyllactose in IL-1ß-treated chondrocytes. Additionally, IL-1ß induction of Erk phosphorylation and IκB degradation, representing a critical signal pathway for osteoarthritic development, was totally blocked by 3'-sialyllactose in a dose-dependent manner. In vivo, 3'-sialyllactose protected against osteoarthritic cartilage destruction in an osteoarthritis mouse model induced by destabilization of the medial meniscus, as demonstrated by histopathological analysis. Our results strongly suggest that 3'-sialyllactose may ameliorate osteoarthritic cartilage destruction by cartilage regeneration via promoting Col2a1 production and may inhibit cartilage degradation and inflammation by suppressing Mmp3, Mmp13 and Cox2 expression. The effects of 3'-sialyllactose could be attributed in part to its regulation of Sox9 or NF-κB and inhibition of Erk phosphorylation and IκB degradation. Taken together, these effects indicate that 3'-sialyllactose merits consideration as a natural therapeutic agent for protecting against osteoarthritis.


Asunto(s)
Cartílago Articular/patología , Homeostasis , Oligosacáridos/uso terapéutico , Osteoartritis/tratamiento farmacológico , Administración Oral , Animales , Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/enzimología , Condrocitos/patología , Colágeno Tipo II/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Meniscos Tibiales/patología , Ratones , FN-kappa B/metabolismo , Oligosacáridos/administración & dosificación , Oligosacáridos/farmacología , Osteoartritis/patología , Proteoglicanos/metabolismo , Factor de Transcripción SOX9/metabolismo , Sulfatos/metabolismo
3.
Dev Biol ; 405(1): 1-9, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116879

RESUMEN

The sh3bgr (SH3 domain binding glutamate-rich) gene encodes a small protein containing a thioredoxin-like fold, SH3 binding domain, and glutamate-rich domain. Originally, it was suggested that increased expression of Sh3bgr may cause the cardiac phenotypes in Down's syndrome. However, it was recently reported that the overexpression of Sh3bgr did not cause any disease phenotypes in mice. In this study, we have discovered that Sh3bgr is critical for sarcomere formation in striated muscle tissues and also for heart development. Sh3bgr is strongly expressed in the developing somites and heart in Xenopus. Morpholino mediated-knockdown of sh3bgr caused severe malformation of heart tissue and disrupted segmentation of the somites. Further analysis revealed that Sh3bgr specifically localized to the Z-line in mature sarcomeres and that knockdown of Sh3bgr completely disrupted sarcomere formation in the somites. Moreover, overexpression of Sh3bgr resulted in abnormally discontinues thick firmaments in the somitic sarcomeres. We suggest that Sh3bgr does its function at least partly by regulating localization of Enah for the sarcomere formation. In addition, we provide the data supporting Sh3bgr is also necessary for proper heart development in part by affecting the Enah protein level.


Asunto(s)
Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Sarcómeros/metabolismo , Tiorredoxinas/química , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Animales , Embrión no Mamífero/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Desarrollo de Músculos , Músculo Estriado/embriología , Músculo Estriado/metabolismo , Miocardio/metabolismo , Estructura Secundaria de Proteína , Transporte de Proteínas , Somitos/embriología , Somitos/metabolismo , Tiorredoxinas/metabolismo , Xenopus/embriología
4.
J Biol Chem ; 286(52): 44480-90, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22033928

RESUMEN

The ADP-ribosyl cyclase CD38 whose catalytic domain resides in outside of the cell surface produces the second messenger cyclic ADP-ribose (cADPR) from NAD(+). cADPR increases intracellular Ca(2+) through the intracellular ryanodine receptor/Ca(2+) release channel (RyR). It has been known that intracellular NAD(+) approaches ecto-CD38 via its export by connexin (Cx43) hemichannels, a component of gap junctions. However, it is unclear how cADPR extracellularly generated by ecto-CD38 approaches intracellular RyR although CD38 itself or nucleoside transporter has been proposed to import cADPR. Moreover, it has been unknown what physiological stimulation can trigger Cx43-mediated export of NAD(+). Here we demonstrate that Cx43 hemichannels, but not CD38, import cADPR to increase intracellular calcium through RyR. We also demonstrate that physiological stimulation such as Fcγ receptor (FcγR) ligation induces calcium mobilization through three sequential steps, Cx43-mediated NAD(+) export, CD38-mediated generation of cADPR and Cx43-mediated cADPR import in J774 cells. Protein kinase A (PKA) activation also induced calcium mobilization in the same way as FcγR stimulation. FcγR stimulation-induced calcium mobilization was blocked by PKA inhibition, indicating that PKA is a linker between FcγR stimulation and NAD(+)/cADPR transport. Cx43 knockdown blocked extracellular cADPR import and extracellular cADPR-induced calcium mobilization in J774 cells. Cx43 overexpression in Cx43-negative cells conferred extracellular cADPR-induced calcium mobilization by the mediation of cADPR import. Our data suggest that Cx43 has a dual function exporting NAD(+) and importing cADPR into the cell to activate intracellular calcium mobilization.


Asunto(s)
Calcio/metabolismo , Conexina 43/metabolismo , ADP-Ribosa Cíclica/metabolismo , NAD/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Transporte Biológico Activo/fisiología , Conexina 43/genética , ADP-Ribosa Cíclica/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , NAD/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
5.
Genes Genomics ; 44(4): 405-413, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35066808

RESUMEN

BACKGROUND: Cell migration is a basic cellular behavior involved in multiple phenomena in the human body such as embryonic development, wound healing, immune reactions, and cancer metastasis. For proper cell migration, integrin and the ECM binding complex must be disassembled for the retraction of trailing edges. OBJECTIVE: Integrin must be differentially regulated at leading edges or trailing edges during cell migration. Previously, we showed that ITGBL1 was a secreted protein and inhibits integrin activity. Therefore, we examined the function of ITGBL1 on the retraction of trailing edges during cell migration. METHODS: To examined the function of ITGBL1 on cell migration, we knocked-down or overexpressed ITGBL1 by using ITGBL1 siRNA or ITGBL1 plasmid DNA in human chondrocytes or ATDC5 cells. We then characterized cellular migration and directionality by performing wound healing assays. Also, to analyze leading-edge formation and trailing-edge retraction, we labeled cell membranes with membrane-GFP and performed live imaging of migrating cells and. Finally, we specifically detected active forms of integrin, FAK and Vinculin using specific antibodies upon ITGBL1 depletion or overexpression. RESULT: In this study, ITGBL1 preferentially inhibited integrin activity at the trailing edges to promote cell migration. ITGBL1-depleted cells showed increased focal adhesions at the membranous traces of trailing edges to prevent the retraction of trailing edges. In contrast, overexpression of ITGBL1 upregulated directional cell migration by promoting focal adhesion disassembly at the trailing edges. CONCLUSION: ITGBL1 facilitates directional cell migration by promoting disassembly of the trailing edge focal adhesion complex.


Asunto(s)
Matriz Extracelular , Adhesiones Focales , Integrina beta1 , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo
6.
Sci Adv ; 8(3): eabl4222, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35061535

RESUMEN

Chondrocytes secrete massive extracellular matrix (ECM) molecules that are produced, folded, and modified in the endoplasmic reticulum (ER). Thus, the ER-associated degradation (ERAD) complex-which removes misfolded and unfolded proteins to maintain proteostasis in the ER- plays an indispensable role in building and maintaining cartilage. Here, we examined the necessity of the ERAD complex in chondrocytes for cartilage formation and maintenance. We show that ERAD gene expression is exponentially increased during chondrogenesis, and disruption of ERAD function causes severe chondrodysplasia in developing embryos and loss of adult articular cartilage. ERAD complex malfunction also causes abnormal accumulation of cartilage ECM molecules and subsequent chondrodysplasia. ERAD gene expression is decreased in damaged cartilage from patients with osteoarthritis (OA), and disruption of ERAD function in articular cartilage leads to cartilage destruction in a mouse OA model.

7.
BMB Rep ; 53(10): 491-499, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32731916

RESUMEN

The extracellular matrix is a critical component of every human tissue. ECM not only functions as a structural component but also regulates a variety of cellular processes such as cell migration, differentiation, proliferation, and cell death. In addition, current studies suggest that ECM is critical for the pathophysiology of various human diseases. ECM is composed of diverse components including several proteins and polysaccharide chains such as chondroitin sulfate, heparan sulfate, and hyaluronic acid. Each component of ECM exerts its own functions in cellular and pathophysiological processes. One of the interesting recent findings is that ECM is involved in inflammatory responses in various human tissues. In this review, we summarized the known functions of ECM in neuroinflammation after acute injury and chronic inflammatory diseases of the central nerve systems. [BMB Reports 2020; 53(10): 491-499].


Asunto(s)
Matriz Extracelular/inmunología , Neuroinmunomodulación/fisiología , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Glicosaminoglicanos , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Neuroinmunomodulación/inmunología , Proteoglicanos
8.
Nutr Res ; 70: 60-69, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30446253

RESUMEN

Studies have highlighted the association between the cellular damage caused by reactive oxygen species and aging. The reducing sugar d-galactose causes aging-related changes and oxidative stress. Lipids are the first target of free radicals, and lipid peroxidation is related to aging. Walnut (Juglans regia Chandler) kernel contains antioxidant phenolic compounds, and chokeberry (Aronia melanocarpa) is one of the richest sources of polyphenols, including anthocyanins, among other fruits. Polyphenols from chokeberry exhibit antioxidant and anti-inflammatory activities. In this study, the additive antioxidative effect of walnut and chokeberry mixture was evaluated by oxidative stress index in d-galactose-induced aging model. Thirty-five Balb/c mice (8 weeks old) were divided into following five groups (n = 7 in each group): normal control (C), d-galactose control (D), d-galactose with chokeberry diet (CH), d-galactose with walnut diet (W), and d-galactose with walnut and chokeberry mixture diet (WCH). In all treatment diets groups, the levels of serum, hepatic, and kidney malonaldehyde were significantly lower than D group and the levels were approaching to control level. Moreover, the kidney malondialdehyde levels were significantly lower in WCH group compared with the control group. This study also confirmed the activities of antioxidant enzymes in liver, as the levels of superoxide dismutase, and glutathione peroxidase were significantly increased in CH group compared to in W or CH groups. The results of this study supported the additive effect of walnut and chokeberry on increment of antioxidant enzyme gene expression in liver and consequently the attenuation of lipid peroxidation in serum, liver, and kidney in d-galactose-induced aging-mouse model. Further studies are needed to investigate the detailed mechanism underlying the additive antioxidative effects in various tissues.


Asunto(s)
Envejecimiento/fisiología , Antioxidantes/farmacología , Galactosa/efectos adversos , Juglans/química , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Photinia/química , Animales , Antocianinas/farmacología , Antioxidantes/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Frutas , Glutatión Peroxidasa/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos BALB C , Nueces , Fenoles/farmacología , Preparaciones de Plantas/farmacología , Polifenoles/farmacología , Semillas , Superóxido Dismutasa/metabolismo
9.
Biochem Biophys Res Commun ; 367(1): 156-61, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18166151

RESUMEN

NAD is available in the extracellular environment and elicits immune modulation such as T cell apoptosis by being used as the substrate of cell surface ADP-ribosyl transferase. However, it is unclear whether extracellular NAD affects function of macrophages expressing cell surface ADP-ribosyl transferase. Here we show that extracellular NAD enhances Fcgamma receptor (FcgammaR)-mediated phagocytosis in J774A.1 macrophages via the conversion into cyclic ADP-ribose (cADPR), a potent calcium mobilizer, by CD38, an ADP-ribosyl cyclase. Extracellular NAD increased the phagocytosis of IgG-coated sheep red blood cells (IgG-SRBC) in J774A.1 macrophages, which was completely abolished by pretreatment of 8-bromo-cADPR, an antagonist of cADPR, or CD38 knockdown. Extracellular NAD increased basal intracellular Ca(2+) concentration, which also was abolished by pretreatment of 8-bromo-cADPR or CD38 knockdown. Moreover, the chelation of intracellular calcium abolished NAD-induced enhancement of phagocytosis of IgG-SRBC. Our results suggest that extracellular NAD act as a regulator for FcgammaR-mediated phagocytosis in macrophages.


Asunto(s)
Calcio/metabolismo , Espacio Extracelular/metabolismo , Macrófagos/fisiología , NAD/metabolismo , Fagocitosis/fisiología , Receptores de IgG/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Transporte Biológico , Células Cultivadas , ADP-Ribosa Cíclica/metabolismo , Inmunoglobulina G/sangre , Macrófagos/patología , Ratones
10.
Biochem Biophys Res Commun ; 377(1): 242-7, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18835379

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) ligands have been identified as a potential source of therapy for human cancers. However, PPARgamma ligands have a limitation for breast cancer therapy, since estrogen receptor alpha (ER(alpha)) negatively interferes with PPARgamma signaling in breast cancer cells. Here we show that ER(alpha) inhihits PPARgamma transactivity and ER(alpha)-mediated inhibition of PPARgamma transactivity is blocked by tamoxifen, an estrogen receptor blocker. The activation of ER(alpha) with 17-beta-estradiol blocked PPRE transactivity induced by troglitazone, a PPARgamma ligand, indicating the resistance of ER(alpha)-positive breast cancer cells to troglitazone. Indeed, troglitazone inhibited the growth of ER(alpha)-negative MDA-MB-231 cells more than that of ER(alpha)-positive MCF-7 cells. Combination of troglitazone with tamoxifen led to a marked increase in growth inhibition of ER(alpha)-positive MCF-7 cells compared to either agent alone. Our data indicates that troglitazone enhances the growth inhibitory activity of tamoxifen in ER(alpha)-positive MCF-7 cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Cromanos/farmacología , Antagonistas de Estrógenos/farmacología , Tamoxifeno/farmacología , Tiazolidinedionas/farmacología , Apoptosis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Estradiol/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Fase G1/efectos de los fármacos , Humanos , Ligandos , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Troglitazona
11.
Int J Hematol ; 87(2): 189-194, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18256785

RESUMEN

The differentiation of promyelocytic leukemic cells into mature cells is the major strategy for drug-based treatment of leukemia. Higher efficient methods to differentiate promyelocytic leukemic cells have been developed using various differentiation inducers including interferon-alpha, interleukin-4, tumor necrosis factor-alpha (TNF-alpha), and dimethyl sulfoxide (DMSO) as a single agent or in combination with each other. Here, we show that a combination of TNF-alpha with DMSO shows a synergic effect on HL-60 cell differentiation through the activation of ERK pathway. TNF-alpha enhanced CD11b expression and percent of cell population in the G1 phase induced by DMSO, which are hallmarks for HL-60 cell differentiation. Inhibition of ERK pathway abolished the synergic effect of TNF-alpha in combination with DMSO on HL-60 differentiation, but the inhibition NF-kappaB pathway did not. These results suggest that TNF-alpha synergistically increases DMSO-induced differentiation of HL-60 cells through the activation of ERK/MAPK-signaling pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dimetilsulfóxido/farmacología , Células Precursoras de Granulocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Sinergismo Farmacológico , Quimioterapia Combinada , Células HL-60 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Fosforilación/efectos de los fármacos
12.
Sci Transl Med ; 10(462)2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305454

RESUMEN

Developing and mature chondrocytes constantly interact with and remodel the surrounding extracellular matrix (ECM). Recent research indicates that integrin-ECM interaction is differentially regulated during cartilage formation (chondrogenesis). Integrin signaling is also a key source of the catabolic reactions responsible for joint destruction in both rheumatoid arthritis and osteoarthritis. However, we do not understand how chondrocytes dynamically regulate integrin signaling in such an ECM-rich environment. Here, we found that developing chondrocytes express integrin-ß-like 1 (Itgbl1) at specific stages, inhibiting integrin signaling and promoting chondrogenesis. Unlike cytosolic integrin inhibitors, ITGBL1 is secreted and physically interacts with integrins to down-regulate activity. We observed that Itgbl1 expression was strongly reduced in the damaged articular cartilage of patients with osteoarthritis (OA). Ectopic expression of Itgbl1 protected joint cartilage against OA development in the destabilization of the medial meniscus-induced OA mouse model. Our results reveal ITGBL1 signaling as an underlying mechanism of protection against destructive cartilage disorders and suggest the potential therapeutic utility of targeting ITGBL1 to modulate integrin signaling in human disease.


Asunto(s)
Cartílago Articular/metabolismo , Cartílago Articular/patología , Condrogénesis , Integrina beta1/metabolismo , Osteoartritis/metabolismo , Osteoartritis/prevención & control , Anciano , Animales , Diferenciación Celular , Línea Celular Tumoral , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Matriz Extracelular/metabolismo , Cara/embriología , Regulación de la Expresión Génica , Humanos , Articulaciones/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Persona de Mediana Edad , Osteoartritis/genética , Osteoartritis/patología , Xenopus/embriología
13.
Int J Biochem Cell Biol ; 39(6): 1260-75, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17521952

RESUMEN

Diabetes mellitus is characterized by cytokine-induced insulitis and a deficit in beta-cell mass. Ligands for peroxisome proliferator-activated receptor-gamma (PPAR-gamma) have been shown to have anti-inflammatory effects in various experimental models. We questioned whether activation of endogenous PPAR-gamma by either PPAR-gamma ligands or adenoviral-directed overexpression of PPAR-gamma (Ad-PPAR-gamma) could inhibit cytokine-induced beta-cell death in RINm5F (RIN) cells, a rat insulinoma cell line. Treatment of RIN cells with interleukin-1 beta (IL-1 beta) and interferon-gamma (IFN-gamma) induced beta-cell damage through NF kappaB-dependent signaling pathways. Activation of PPAR-gamma by PPAR-gamma ligands or Ad-PPAR-gamma inhibited IL-1 beta and IFN-gamma-stimulated nuclear translocation of the p65 subunit and DNA binding activity. NF kappaB target gene expression and their product formation, namely inducible nitric oxide synthase and cyclooxygenase-2 were decreased by PPAR-gamma activation, as established by real-time PCR, Western blots and measurements of NO and PGE(2). The mechanism by which PPAR-gamma activation inhibited NF kappaB-dependent cell death signals appeared to involve the inhibition of I kappa B alpha degradation, evidenced by inhibition of cytokine-induced NF kappaB-dependent signaling events by Ad-I kappaB alpha (S32A, S36A), non-degradable I kappaB alpha mutant. I kappaB beta mutant, Ad-I kappaB beta (S19A, S23A) was not effective in preventing cytokine toxicity. Furthermore, a protective effect of PPAR-gamma ligands was proved by assaying for normal insulin secreting capacity in response to glucose in isolated rat pancreatic islets. The beta-cell protective function of PPAR-gamma ligands might serve to counteract cytokine-induced beta-cell destruction.


Asunto(s)
Citocinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Adenoviridae/genética , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromanos/farmacología , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacología , Masculino , Mutación , Inhibidor NF-kappaB alfa , Nitritos/metabolismo , PPAR gamma/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología , Troglitazona
14.
Exp Mol Med ; 39(1): 121-7, 2007 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-17334236

RESUMEN

TNF-alpha plays a variety of biological functions such as apoptosis, inflammation and immunity. PTEN also has various cellular function including cell growth, proliferation, migration and differentiation. Thus, possible relationships between the two molecules are suggested. TNF-alpha has been known to downregulate PTEN via NF-kappaB pathway in the human colon cell line, HT-29. However, here we show the opposite finding that TNF-alpha upregulates PTEN via activation of NF-kappaB in human leukemic cells. TNF-alpha increased PTEN expression at HL-60 cells in a time- and dose-dependent manner, but the response was abolished by disruption of NF-kappaB with p65 antisense phosphorothioate oligonucleotide or pyrrolidine dithiocarbamate. We found that TNF-alpha activated the NF-kappaB pathways, evidenced by the translocation of p65 to the nucleus in TNF-alpha-treated cells. We conclude that TNF-alpha induces upregulation of PTEN expression through NF-kappaB activation in human leukemic cells.


Asunto(s)
Leucemia/metabolismo , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Línea Celular Tumoral , Expresión Génica , Humanos , Leucemia/genética , FN-kappa B/genética , Fosfohidrolasa PTEN/genética
15.
Exp Mol Med ; 39(5): 673-8, 2007 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18059143

RESUMEN

Lipoprotein plays a role in the host defense against bacterial infection, and its serum level has been demonstrated to be an important prognosis factor of survival. We have previously demonstrated that LDL directly inactivates the hemolytic activity of Vibrio vulnificus cytolysin (VVC) in vitro. The object of this study was therefore to examine whether the LDL-mediated inactivation of VVC leads to protection against lethal infection of V. vulnificus in vivo, using wild and VVC-deficient V. vulnificus strains. Unexpectedly, we found that LDL protects mouse lethality induced by VVC-deficient as well as wild V. vulnificus strain. We also demonstrated that LDL blocks V. vulnificus LPS-induced lethality in mice. These results suggest that LDL preferentially act on endotoxin rather than exotoxin in the protection against V. vulnificus-induced mice lethality.


Asunto(s)
Lipopolisacáridos/antagonistas & inhibidores , Lipoproteínas LDL/farmacología , Vibrio vulnificus/efectos de los fármacos , Vibrio vulnificus/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Perforina/antagonistas & inhibidores , Perforina/genética , Vibriosis/prevención & control , Vibrio vulnificus/genética , Virulencia/efectos de los fármacos , Virulencia/genética , Virulencia/fisiología
16.
Int J Hematol ; 85(3): 231-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17483060

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) and retinoic acid receptors (RARs) have been a focus in chemotherapy for human cancers. The tumor suppressor PTEN plays a pivotal role in the growth of human cancer cells. We investigated whether costimulation of PPARgamma and RAR could synergistically up-regulate PTEN in human leukemia cells and consequently potentiate the inhibition of growth and cell cycle progression of these cells. We found that overexpression of PTEN with the adenoviral vector Ad/PTEN caused growth arrest at the G1 phase of the cell cycle of HL-60 cells. HL-60 cells treated with either a PPARgamma ligand (ciglitazone) or a RAR ligand (all-trans retinoic acid [ATRA]) up-regulated PTEN in HL-60 cells. The 2 compounds in combination showed synergistic effects on PTEN expression at the protein and messenger RNA levels. Moreover, the combination of ciglitazone and ATRA synergistically reduced cell growth rates and cell cycle arrest at the G1 phase. Our results suggest that, PPARgamma and RAR play an important role in controlling the growth of leukemia cells via the up-regulation of PTEN.


Asunto(s)
Ciclo Celular/fisiología , PPAR gamma/fisiología , Fosfohidrolasa PTEN/metabolismo , Receptores de Ácido Retinoico/fisiología , Células HL-60 , Humanos , Tiazolidinedionas/farmacología , Tretinoina/farmacología , Regulación hacia Arriba
17.
Clin Infect Dis ; 43(5): 585-92, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16886150

RESUMEN

BACKGROUND: This study was performed to evaluate the associations of newly recognized viruses, namely, human metapneumovirus (hMPV), human coronavirus (HCoV)-NL63, and human bocavirus (HBoV) with lower respiratory tract infections (LRTIs) in previously healthy children. METHODS: To determine the prevalences of 11 viruses--respiratory syncytial virus (RSV), adenovirus, rhinovirus, parainfluenza viruses (PIVs) 1 and 3, influenza viruses A and B, hMPV, HCoV, HCoV-NL63, and HBoV--among infants or children with LRTIs, in association with their epidemiologic characteristics, we performed multiplex reverse-transcriptase polymerase chain reaction on nasopharyngeal aspirates obtained from 515 children < or =5 years old with LRTIs during the period 2000-2005. RESULTS: Viruses were identified in 312 (60.6%) of the 515 patients. RSV was detected in 122 (23.7%), HBoV in 58 (11.3%), adenovirus in 35 (6.8%), PIV-3 in 32 (6.2%), rhinovirus in 30 (5.8%), hMPV in 24 (4.7%), influenza A in 24 (4.7%), PIV-1 in 9 (1.7%), influenza B in 9 (1.7%), and HCoV-NL63 in 8 (1.6%). Coinfections with > or =2 viruses were observed in 36 patients (11.5%). Twenty-two patients (37.9%) infected with HBoV had a coinfection. Bronchiolitis was frequently diagnosed in patients who tested positive for RSV, PIV-3, or rhinovirus, whereas influenza A, PIV-1, and HCoV-NL63 were commonly found in patients with croup. The age distributions of patients with viral infections differed; notably, RSV was responsible for 77% of LRTIs that occurred in infants < or =3 months old. The number of hMPV infections peaked between February and April, whereas the number of HCoV-NL63 infections peaked between April and May. CONCLUSIONS: This study describes the features of LRTIs associated with newly identified viruses in children, compared with those associated with known viruses. Additional investigations are required to define the role of HBoV in LRTI.


Asunto(s)
Virosis/epidemiología , Virosis/virología , Virus/clasificación , Virus/aislamiento & purificación , Preescolar , Humanos , Lactante , Corea (Geográfico)/epidemiología
18.
J Clin Virol ; 35(4): 388-93, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16458589

RESUMEN

BACKGROUND: Adenovirus type 7 (Ad7) is frequently responsible for severe respiratory infections, especially in young infants. Since the Ad7 epidemics have been associated with severe childhood pneumonia and significant mortality in Korea, 1995-1999, continuous surveillance was necessary for Ad7 related diseases. OBJECTIVES: To characterize epidemiologic features of Ad7 in 1995-2004, genetic diversity of Ad7 were studied by determining genome types (GTs) and the fiber diversity. STUDY DESIGN: A total of 139 Ad7 strains were obtained from Korean children with pneumonia. Serotype specificity was confirmed by microneutralization assay. GTs were determined by restriction analysis with 12 enzymes. The variable region of the fiber was sequenced. RESULTS: Two GTs, Ad7d (N=98, 71%) and Ad7l (N=41, 29%) have been identified. In 1995-1996 and 2001-2002, Ad7d was dominant accounting for 98-100% of all Ad7; in 1999-2000, Ad7l was the prevalent GT accounting for 100% of all Ad7; in 1997-1998 and 2003-2004, both GTs circulated concurrently. The change in the relative predominance of GT occurred in 2 or 3 years. The Lys substitution for Arg at codon 280 of the fiber was identified in 31 Ad7d strains (32%) while no variations were observed among Ad7l. It was noteworthy that two fiber variants of Ad7d were not concurrently prevalent on any time after 1996. The shift in predominant fiber variants of Ad7d was also observed in 2-3 years. CONCLUSIONS: Our data demonstrated that the two GTs, Ad7d and Ad7l circulated in an alternating manner between outbreaks of Ad7 associated childhood pneumonia over 10 consecutive years in Korea. Fiber diversity at position 280 within Ad7d appeared to contribute to the annual distribution of Ad7d. This observation necessitates further studies to demonstrate an association between fiber variation and host cell specificity or neutralization antibody recognition.


Asunto(s)
Infecciones por Adenovirus Humanos/epidemiología , Adenovirus Humanos/genética , Proteínas de la Cápside/genética , Variación Genética , Epidemiología Molecular , Neumonía Viral/epidemiología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/clasificación , Adenovirus Humanos/aislamiento & purificación , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de la Cápside/química , Línea Celular , Niño , Preescolar , Humanos , Corea (Geográfico)/epidemiología , Neumonía Viral/virología
19.
Sci Rep ; 6: 24159, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27063487

RESUMEN

We construct a novel recombinant secondary antibody mimic, GST-ABD, which can bind to the Fc regions of target-bound primary antibodies and acquire multiple HRPs simultaneously. We produce it in tenth of mg quantities with a bacterial overexpression system and simple purification procedures, significantly reducing the manufacturing cost and time without the use of animals. GST-ABD is effectively conjugated with 3 HRPs per molecule on an average and selectively bind to the Fc region of primary antibodies derived from three different species (mouse, rabbit, and rat). HRP-conjugated GST-ABD (HRP-GST-ABD) is successfully used as an alternative to secondary antibodies to amplify target-specific signals in both ELISA and immunohistochemistry regardless of the target molecules and origin of primary antibodies used. GST-ABD also successfully serves as an anchoring adaptor on the surface of GSH-coated plates for immobilizing antigen-capturing antibodies in an orientation-controlled manner for sandwich-type indirect ELISA through simple molecular recognition without any complicated chemical modification.


Asunto(s)
Anticuerpos/inmunología , Ensayo de Inmunoadsorción Enzimática , Animales , Glutatión/química , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Peroxidasa de Rábano Silvestre/química , Peroxidasa de Rábano Silvestre/genética , Peroxidasa de Rábano Silvestre/metabolismo , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunohistoquímica , Cinética , Ratones , Tecnicas de Microbalanza del Cristal de Cuarzo , Conejos , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Resonancia por Plasmón de Superficie
20.
Sci Rep ; 6: 20261, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-27064360

RESUMEN

Terminally misfolded proteins are selectively recognized and cleared by the endoplasmic reticulum-associated degradation (ERAD) pathway. SEL1L, a component of the ERAD machinery, plays an important role in selecting and transporting ERAD substrates for degradation. We have determined the crystal structure of the mouse SEL1L central domain comprising five Sel1-Like Repeats (SLR motifs 5 to 9; hereafter called SEL1L(cent)). Strikingly, SEL1L(cent) forms a homodimer with two-fold symmetry in a head-to-tail manner. Particularly, the SLR motif 9 plays an important role in dimer formation by adopting a domain-swapped structure and providing an extensive dimeric interface. We identified that the full-length SEL1L forms a self-oligomer through the SEL1L(cent) domain in mammalian cells. Furthermore, we discovered that the SLR-C, comprising SLR motifs 10 and 11, of SEL1L directly interacts with the N-terminus luminal loops of HRD1. Therefore, we propose that certain SLR motifs of SEL1L play a unique role in membrane bound ERAD machinery.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Proteínas/química , Proteínas/metabolismo , Secuencias de Aminoácidos , Animales , Cristalografía por Rayos X , Péptidos y Proteínas de Señalización Intracelular , Ratones , Conformación Proteica , Secuencias Repetitivas de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA