Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Biochem Biophys Res Commun ; 526(4): 1061-1068, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32312517

RESUMEN

Persistent infection with high-risk strains of human papillomavirus (HPV) is the primary cause of cervical cancer, the fourth most common cancer among women worldwide. Two oncoproteins encoded by the HPV genome, E6 and E7, are required for epigenetic modifications that promote cervical cancer development. We found that knockdown of HPV E6/E7 by siRNA reduced the levels of ubiquitin-like containing PHD and RING finger domain 1 (UHRF1) but increased the levels of gelsolin (GSN) in early stage cervical cancer cells. In addition, we found that UHRF1 levels were increased and GSN levels were decreased in early stage cervical cancer compared with those in normal cervical tissues, as shown by Western blot analysis, immunohistochemistry, and analysis of the Oncomine database. Moreover, knockdown of UHRF1 resulted in increased cell death in cervical cancer cell lines. Treatment of E6/E7-transformed HaCaT (HEK001) cells and HeLa cells with the DNA-hypomethylating agent 5-aza-2'-deoxycytidine and the histone deacetylase inhibitor Trichostatin A increased GSN expression levels. UHRF1 knockdown in HEK001 cells by siRNA or the UHRF1 antagonist thymoquinone increased GSN levels, induced cell cycle arrest and apoptosis, and increased the levels of p27 and cleaved PARP. Those results indicate that upregulation of UHRF1 by HPV E6/E7 causes GSN silencing and a reduction of cell death in early stage cervical cancer, suggesting that GSN might be a useful therapeutic target in early stage cervical cancer.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Gelsolina/metabolismo , Silenciador del Gen , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Adulto , Anciano , Anciano de 80 o más Años , Benzoquinonas/farmacología , Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , Puntos de Control del Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas E7 de Papillomavirus/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
2.
Biochem Biophys Res Commun ; 503(3): 1307-1314, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30017190

RESUMEN

Oxidative stress plays an important role in the development of diabetic retinopathy. Here, we examined whether α-lipoic acid (α-LA), a natural antioxidant, attenuated retinal injury in diabetic mice. The α-LA was orally administered to control mice or mice with streptozotocin-induced diabetes. We found that α-LA reduced oxidative stress, decreased and increased retinal 4-hydroxy-2-nonenal and glutathione peroxidase, respectively, and inhibited retinal cell death. Concomitantly, α-LA reversed the decreased activation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase, and increased the levels of peroxisome proliferator-activated receptor delta and sirtuin3 in diabetic mouse retinas, similar to results shown after metformin treatment of retinal pigment epithelial cells (RPE) exposed to high glucose. Moreover, α-LA lowered the levels of O-linked ß-N-acetylglucosamine transferase (OGT) and thioredoxin-interacting protein (TXNIP) in diabetic retinas that were more pronounced after metformin treatment of RPE cells. Importantly, α-LA lowered interactions between AMPK and OGT as shown by co-immunoprecipitation analyses, and this was accompanied by less cell death as measured by double immunofluorescence staining by terminal deoxynucleotide transferase-mediated dUTP nick-end labelling and OGT or TXNIP in retinal ganglion cells. Consistently, α-LA lowered the levels of cleaved poly(ADP-ribose) polymerase and pro-apoptotic marker cleaved caspase-3 in diabetic retinas. Our results indicated that α-LA reduced retinal cell death partly through AMPK activation or OGT inhibition in diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Retina/citología , Retina/efectos de los fármacos , Ácido Tióctico/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Administración Oral , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/metabolismo , Retina/metabolismo , Retina/patología , Estreptozocina , Ácido Tióctico/administración & dosificación
3.
Biochem Biophys Res Commun ; 492(3): 397-403, 2017 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-28843855

RESUMEN

Retinal degeneration is an early feature of diabetic retinopathy, the major cause of blindness in the developed world. Here we investigated how the widely used antidiabetic drug metformin reduces retinal injury in diabetic mice. Metformin was orally administered to control mice or mice with streptozotocin-induced diabetes. Western blot analysis showed that levels of O-linked ß-N-acetylglucosamine (O-GlcNAc) transferase (OGT) and other related proteins such as carbohydrate-responsive element-binding protein (ChREBP) and thioredoxin-interacting protein (TXNIP) were significantly increased, and nuclear factor kappaB (NF-κB) and poly (ADP-ribose) polymerase (PARP) were activated in the diabetic retinas or retinal pigment epithelial (RPE) cells exposed to high glucose compared to controls. More importantly, RPE cells exposed to high glucose and treated with thiamet-G had higher levels of those proteins, demonstrating the role of elevated O-GlcNAcylation. Double immunofluorescence analysis revealed increased co-localization of terminal deoxynucleotide transferase-mediated dUTP nick-end labelling (TUNEL)-positive ganglion cells and OGT, ChREBP, TXNIP, or NF-κB in diabetic retinas compared to control retinas. Co-immunoprecipitation analysis showed that interaction between OGT and ChREBP or NF-κB was increased in diabetic retinas compared to control retinas, and this was accompanied by more cell death. Notably, metformin attenuated the increases in protein levels; reduced co-localization of TUNEL-positive ganglion cells and OGT, ChREBP, TXNIP, or NF-κB; and reduced interaction between OGT and ChREBP or NF-κB. Our results indicate that OGT inhibition might be one of the mechanisms by which metformin decreases retinal cell death.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Hipoglucemiantes/farmacología , Metformina/farmacología , Retina/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Humanos , Hipoglucemiantes/administración & dosificación , Masculino , Metformina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Retina/citología , Retina/patología , Estreptozocina , Aumento de Peso/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 483(2): 793-802, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-27845045

RESUMEN

High-risk human papilloma virus (HPV) 16/18 infections are often found in lung cancer. The cellular mechanisms involved in the metastatic spread of HPV-infected cervical cancer cells remain largely elusive. High O-linked-N-acetylglucosamine (O-GlcNAc) modification has also been observed in lung cancer. In the present study, we assessed the relationship between O-GlcNAc transferase (OGT) and HPV 16/18 E6/E7, or C-X-C chemokine receptor type 4 (CXCR4), in HeLa cells and in lungs of xenografted mice. Depleting OGT with an OGT-specific shRNA significantly decreased levels of E6 and E7 oncoproteins in HeLa cells and xenograft tumors, and reduced tumor formation in vivo. Western blotting and immunofluorescence analysis showed significantly decreased expression levels of E6, E7, and HCF-1 in the lungs of xenografted mice treated with an OGT-specific shRNA compared to those treated with non-targeting shRNA. Additionally, levels of E7 or OGT co-localized with Ki-67 were significantly decreased in the lungs of xenografted mice treated with OGT-specific shRNA compared to those treated with non-targeting shRNA. Moreover, levels of CXCR4 were significantly decreased in HeLa cells and in the lungs of xenografted mice treated with OGT-specific shRNA compared to those treated with non-targeting shRNA; this may be related to reduced adhesion or invasion of circulating HPV-positive tumor cells. These findings provide novel evidence that OGT functions in metastatic spread of HPV E6/E7-positive tumor cells to the lungs through E6/E7, HCF-1 and CXCR4, suggesting OGT might be a therapeutic target for HPV-positive lung cancer.


Asunto(s)
Papillomavirus Humano 16/patogenicidad , Papillomavirus Humano 18/patogenicidad , Neoplasias Pulmonares/etiología , N-Acetilglucosaminiltransferasas/metabolismo , Infecciones por Papillomavirus/etiología , Animales , Proteínas de Unión al ADN/metabolismo , Células HeLa , Xenoinjertos , Factor C1 de la Célula Huésped/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/virología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , ARN Interferente Pequeño/genética , Receptores CXCR4/metabolismo , Proteínas Represoras/metabolismo
5.
Biochim Biophys Acta ; 1852(7): 1550-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25916635

RESUMEN

Hepatic ischemia/reperfusion (I/R) injury can arise as a complication of liver surgery and transplantation. Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, modulates inflammation and apoptosis in response to oxidative stress. SIRT1, which is regulated by p53 and microRNA-34a (miR-34a), can modulate non-alcoholic fatty liver disease, fibrosis and cirrhosis. Since carbon monoxide (CO) inhalation can protect against hepatic I/R, we hypothesized that CO could ameliorate hepatic I/R injury by regulating the miR-34a/SIRT1 pathway. Livers from mice pretreated with CO, or PFT, a p53 inhibitor, displayed reduced production of pro-inflammatory mediators, including TNF-α, iNOS, interleukin (IL)-6, and IL-1ß after hepatic I/R injury. SIRT1 expression was increased by CO or PFT in the liver after I/R, whereas acetylated p65, p53 levels, and miR-34a expression were decreased. CO increased SIRT1 expression by inhibiting miR-34a. Both CO and PFT diminished pro-inflammatory cytokines production in vitro. Knockdown of SIRT1 in LPS-stimulated macrophages increased NF-κB acetylation, and increased pro-inflammatory cytokines. CO treatment reduced miR-34a expression and increased SIRT1 expression in oxidant-challenged hepatocytes; and rescued SIRT1 expression in p53-expressing or miR-34a transfected cells. In response to CO, enhanced SIRT1 expression mediated by miR-34a inhibition protects against liver damage through p65/p53 deacetylation, which may mediate inflammatory responses and hepatocellular apoptosis. The miR-34a/SIRT1 pathway may represent a therapeutic target for hepatic injury.


Asunto(s)
Monóxido de Carbono/farmacología , Hígado/irrigación sanguínea , MicroARNs/genética , Daño por Reperfusión/metabolismo , Sirtuina 1/metabolismo , Animales , Monóxido de Carbono/uso terapéutico , Línea Celular , Células Cultivadas , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Daño por Reperfusión/prevención & control , Sirtuina 1/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Biochem Biophys Res Commun ; 472(1): 276-80, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26926565

RESUMEN

A healthy acute stress response requires both rapid increase and rapid clearance of blood corticosteroids. We previously showed that regulators of G-protein signaling 4 (RGS4), which decreases in the paraventricular nucleus (PVN) during acute stress, forms a complex with the GABAB receptor. In the present study, we show that this decrease in RGS4 levels in the PVN during an acute stress response facilitates the return of blood corticosteroids to basal levels. Moreover, the effect of RGS4 decrease is attenuated by a GABAB receptor antagonist. These results suggest that RGS4 in the PVN regulates blood corticosteroid-related GABAB receptor signaling during the acute stress response.


Asunto(s)
Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas RGS/metabolismo , Receptores de GABA-B/metabolismo , Estrés Fisiológico , Animales , Corticosterona/sangre , Antagonistas de Receptores de GABA-B/farmacología , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos Organofosforados/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Proteínas RGS/antagonistas & inhibidores , Proteínas RGS/genética , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos
7.
Am J Pathol ; 185(11): 2867-74, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26348577

RESUMEN

Low-dose inhaled carbon monoxide is reported to suppress inflammatory responses and exhibit a therapeutic effect in models of lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, the precise mechanism by which carbon monoxide confers protection against ALI is not clear. Tristetraprolin (TTP; official name ZFP36) exerts anti-inflammatory effects by enhancing decay of proinflammatory cytokine mRNAs. With the use of TTP knockout mice, we demonstrate here that the protection by carbon monoxide against LPS-induced ALI is mediated by TTP. Inhalation of carbon monoxide substantially increased the pulmonary expression of TTP. carbon monoxide markedly enhanced the decay of mRNA-encoding inflammatory cytokines, blocked the expression of inflammatory cytokines, and decreased tissue damage in LPS-treated lung tissue. Moreover, knockout of TTP abrogated the anti-inflammatory and tissue-protective effects of carbon monoxide in LPS-induced ALI. These results suggest that carbon monoxide-induced TTP mediates the protective effect of carbon monoxide against LPS-induced ALI by enhancing the decay of mRNA encoding proinflammatory cytokines.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios/farmacología , Monóxido de Carbono/farmacología , Tristetraprolina/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Citocinas/análisis , Citocinas/genética , Femenino , Regulación de la Expresión Génica , Lipopolisacáridos/efectos adversos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tristetraprolina/efectos de los fármacos , Tristetraprolina/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 309(1): G21-9, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25951827

RESUMEN

Hepatic ischemia-reperfusion (I/R) can cause hepatocellular injury associated with the inflammatory response and mitochondrial dysfunction. We studied the protective effects of the phosphodiesterase inhibitor cilostazol in hepatic I/R and the roles of mitochondria and the Nrf2/heme oxygenase-1 (HO-1) system. Wild-type, Hmox1(-/-), or Nrf2(-/-) mice were subjected to hepatic I/R in the absence or presence of cilostazol followed by measurements of liver injury. Primary hepatocytes were subjected to cilostazol with the HO-1 inhibitor ZnPP, or Nrf2-specific siRNA, followed by assessment of mitochondrial biogenesis. Preconditioning with cilostazol prior to hepatic I/R protected against hepatocellular injury and mitochondrial dysfunction. Cilostazol reduced the serum levels of alanine aminotransferase, TNF-α, and liver myeloperoxidase content relative to control I/R-treated mice. In primary hepatocytes, cilostazol increased the expression of HO-1, and markers of mitochondrial biogenesis, PGC-1α, NRF-1, and TFAM, induced the mitochondrial proteins COX III and COX IV and increased mtDNA and mitochondria content. Pretreatment of primary hepatocytes with ZnPP inhibited cilostazol-induced PGC-1α, NRF-1, and TFAM mRNA expression and reduced mtDNA and mitochondria content. Genetic silencing of Nrf2 prevented the induction of HO-1 and mitochondrial biogenesis by cilostazol in HepG2 cells. Cilostazol induced hepatic HO-1 production and mitochondrial biogenesis in wild-type mice, but not in Hmox1(-/-) or Nrf2(-/-) mice, and failed to protect against liver injury in Nrf2(-/-) mice. These results suggest that I/R injury can impair hepatic mitochondrial function, which can be reversed by cilostazol treatment. These results also suggest that cilostazol-induced mitochondrial biogenesis was mediated by an Nrf-2- and HO-1-dependent pathway.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hígado/irrigación sanguínea , Hígado/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Recambio Mitocondrial/efectos de los fármacos , Sustancias Protectoras/farmacología , Daño por Reperfusión/prevención & control , Tetrazoles/farmacología , Animales , Cilostazol , Citoprotección , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Células Hep G2 , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Hígado/enzimología , Hígado/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Noqueados , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/patología , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Inhibidores de Fosfodiesterasa 3/farmacología , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Korean J Physiol Pharmacol ; 19(5): 451-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26330758

RESUMEN

Sirtuin 1 (SIRT1) is a mammalian NAD(+)-dependent protein deacetylase that regulates cellular metabolism and inflammatory response. The organ-specific deletion of SIRT1 induces local inflammation and insulin resistance in dietary and genetic obesity. Macrophage-mediated inflammation contributes to insulin resistance and metabolic syndrome, however, the macrophage-specific SIRT1 function in the context of obesity is largely unknown. C57/BL6 wild type (WT) or myeloid-specific SIRT1 knockout (KO) mice were fed a high-fat diet (HFD) or normal diet (ND) for 12 weeks. Metabolic parameters and markers of hepatic steatosis and inflammation in liver were compared in WT and KO mice. SIRT1 deletion enhanced HFD-induced changes on body and liver weight gain, and increased glucose and insulin resistance. In liver, SIRT1 deletion increased the acetylation, and enhanced HFD-induced nuclear translocation of nuclear factor kappa B (NF-κB), hepatic inflammation and macrophage infiltration. HFD-fed KO mice showed severe hepatic steatosis by activating lipogenic pathway through sterol regulatory element-binding protein 1 (SREBP-1), and hepatic fibrogenesis, as indicated by induction of connective tissue growth factor (CTGF), alpha-smooth muscle actin (α-SMA), and collagen secretion. Myeloid-specific deletion of SIRT1 stimulates obesity-induced inflammation and increases the risk of hepatic fibrosis. Targeted induction of macrophage SIRT1 may be a good therapy for alleviating inflammation-associated metabolic syndrome.

10.
Blood ; 119(11): 2523-32, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22262759

RESUMEN

The circulating peptide hormone hepcidin maintains systemic iron homeostasis. Hepcidin production increases during inflammation and as a result of endoplasmic reticulum (ER) stress. Elevated hepcidin levels decrease dietary iron absorption and promote iron sequestration in reticuloendothelial macrophages. Furthermore, increased plasma hepcidin levels cause hypoferremia and the anemia associated with chronic diseases. The signal transduction pathways that regulate hepcidin during inflammation and ER stress include the IL-6-dependent STAT-3 pathway and the unfolded protein response-associated cyclic AMP response element-binding protein-H (CREBH) pathway, respectively. We show that carbon monoxide (CO) suppresses hepcidin expression elicited by IL-6- and ER-stress agents by inhibiting STAT-3 phosphorylation and CREBH maturation, respectively. The inhibitory effect of CO on IL-6-inducible hepcidin expression is dependent on the suppressor of cytokine signaling-3 (SOCS-3) protein. Induction of ER stress in mice resulted in increased hepatic and serum hepcidin. CO administration inhibited ER-stress-induced hepcidin expression in vivo. Furthermore, ER stress caused iron accumulation in splenic macrophages, which could be prevented by CO. Our findings suggest novel anti-inflammatory therapeutic applications for CO, as well as therapeutic targets for the amelioration of anemia in the hypoferremic condition associated with chronic inflammatory and metabolic diseases.


Asunto(s)
Antimetabolitos/farmacología , Péptidos Catiónicos Antimicrobianos/antagonistas & inhibidores , Monóxido de Carbono/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inflamación/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Western Blotting , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hepcidinas , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hierro/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
11.
Exp Eye Res ; 122: 13-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24631337

RESUMEN

Recent studies revealed that Tonicity-responsive enhancer binding protein (TonEBP) directly regulates the transcription of aldose reductase (AR), which catalyzes the first step of the polyol pathway of glucose metabolism. Activation of protein kinase C δ (PKCδ) is dependent on AR and it has been linked to diabetic complications. However, whether TonEBP affects expressions of AR and PKCδ in diabetic retinopathy was not clearly shown. In this study, we used TonEBP heterozygote mice to study the role of TonEBP in streptozotocin (STZ)-induced diabetic retinopathy. We performed immunofluorescence staining and found that retinal expressions of AR and PKCδ were significantly reduced in the heterozygotes compared to wild type littermates, particularly in ganglion cell layer. To examine further the effect of TonEBP reduction in retinal tissues, we performed intravitreal injection of TonEBP siRNA and confirmed the decrease in AR and PKCδ levels. In addition, we found that a proapoptotic factor, Bax level was reduced and a survival factor, Bcl2 level was increased after injection of TonEBP siRNA, indicating that TonEBP mediates apoptotic cell death. In parallel, TonEBP siRNA was applied to the in vitro human retinal pigment epithelial (ARPE-19) cells cultured in high glucose media. We have consistently found the decrease in AR and PKCδ levels and changes in apoptotic factors for survival. Together, these results clearly demonstrated that hyperglycemia-induced TonEBP plays a crucial role in increasing AR and PKCδ levels and leading to apoptotic death. Our findings suggest that TonEBP reduction is an effective therapeutic strategy for diabetic retinopathy.


Asunto(s)
Aldehído Reductasa/metabolismo , Retinopatía Diabética/enzimología , Modelos Animales de Enfermedad , Proteína Quinasa C/metabolismo , Factores de Transcripción/fisiología , Animales , Apoptosis , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/enzimología , Retinopatía Diabética/patología , Retinopatía Diabética/prevención & control , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , Retina/enzimología , Células Ganglionares de la Retina/enzimología , Proteína X Asociada a bcl-2/metabolismo
12.
J Psychiatry Neurosci ; 38(3): 183-91, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23031251

RESUMEN

BACKGROUND: The brain levels of glutamate (Glu) and glutamine (Gln) are partially regulated through the Glu-Gln cycle. Astrocytes play a role in regulating the Glu-Gln cycle, and loss of astrocytes has been associated with depressive disorders. We hypothesized that levels of Glu and Gln would be affected by astrocyte loss and dysregulation of the Glu-Gln cycle and that depressive-like behaviours would be closely related to the level of changes in Glu and Gln. METHODS: We used liquid chromatography to measure Glu and Gln concentrations in the prefrontal cortex of male mice infused with L-α aminoadipic acid (L-AAA), a specific astrocyte toxin, in the prelimbic cortex. Methionine sulfoximine, a Gln synthetase inhibitor, and α-methyl-amino-isobutyric acid, a blocker of neuronal Gln transporters, were used to disturb the Glu-Gln cycle. We assessed the behavioural change by drug infusion using the forced swim test (FST) and sucrose preference test. RESULTS: The Glu and Gln levels were decreased on the fifth day after L-AAA infusion, and the infused mice showed longer durations of immobility in the FST and lower sucrose preference, indicative of depressive-like behaviour. Mice in which Gln synthetase or Gln transport were inhibited also exhibited increased immobility in the FST. Direct infusion of L-Gln reversed the increased immobility induced by astrocyte ablation and Glu-Gln cycle impairments. LIMITATIONS: Genetically modified animal models and diverse behavioural assessments would have been helpful to solidify our conclusions. CONCLUSION: Neuronal Gln deficiency in the prefrontal cortex may cause depressive behaviours.


Asunto(s)
Trastorno Depresivo/etiología , Glutamina/deficiencia , Corteza Prefrontal/metabolismo , Ácido 2-Aminoadípico/farmacología , Ácidos Aminoisobutíricos/farmacología , Análisis de Varianza , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Trastorno Depresivo/metabolismo , Inhibidores Enzimáticos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Ácido Glutámico/metabolismo , Glutamina/antagonistas & inhibidores , Masculino , Metionina/análogos & derivados , Metionina/farmacología , Ratones , Ratones Endogámicos C57BL
13.
J Cell Physiol ; 227(3): 1157-67, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21567405

RESUMEN

Clusterin (CLU), a glycoprotein, is involved in apoptosis, producing two alternatively spliced isoforms in various cell types. The pro-apoptotic CLU appears to be a nuclear isoform (nuclear clusterin; nCLU), and the secretory CLU (sCLU) is thought to be anti-apoptotic. The detailed molecular mechanism of nCLU as a pro-apoptotic molecule has not yet been clear. In the current study, overexpressed nCLU induced apoptosis in human kidney cells. Biochemical studies revealed that nCLU sequestered Bcl-XL via a putative BH3 motif in the C-terminal coiled coil (CC2) domain, releasing Bax, and promoted apoptosis accompanied by activation of caspase-3 and cytochrome c release. These results suggest a novel mechanism of apoptosis mediated by nCLU as a pro-apoptotic molecule.


Asunto(s)
Apoptosis , Supervivencia Celular , Clusterina/fisiología , Proteína bcl-X/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Clusterina/química , Clusterina/genética , Células HEK293 , Humanos , Queratinocitos/citología , Queratinocitos/fisiología , Masculino , Datos de Secuencia Molecular , Neoplasias de la Próstata , Dominios y Motivos de Interacción de Proteínas/genética , Estructura Terciaria de Proteína/fisiología , Proteína bcl-X/química , Proteína bcl-X/genética
14.
BMC Neurosci ; 13: 58, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22672618

RESUMEN

BACKGROUND: The amygdala plays an essential role in controlling emotional behaviors and has numerous connections to other brain regions. The functional role of the amygdala has been highlighted by various studies of stress-induced behavioral changes. Here we investigated gene expression changes in the amygdala in the chronic immobilization stress (CIS)-induced depression model. RESULTS: Eight genes were decreased in the amygdala of CIS mice, including genes for neurotrophic factors and extracellular matrix proteins. Among these, osteoglycin, fibromodulin, insulin-like growth factor 2 (Igf2), and insulin-like growth factor binding protein 2 (Igfbp2) were further analyzed for histological expression changes. The expression of osteoglycin and fibromodulin simultaneously decreased in the medial, basolateral, and central amygdala regions. However, Igf2 and Igfbp2 decreased specifically in the central nucleus of the amygdala. Interestingly, this decrease was found only in the amygdala of mice showing higher immobility, but not in mice displaying lower immobility, although the CIS regimen was the same for both groups. CONCLUSIONS: These results suggest that the responsiveness of the amygdala may play a role in the sensitivity of CIS-induced behavioral changes in mice.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Depresión/patología , Regulación hacia Abajo/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Análisis de Varianza , Animales , Condicionamiento Operante/fisiología , Depresión/etiología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Fibromodulina , Preferencias Alimentarias/fisiología , Perfilación de la Expresión Génica , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Factores de Crecimiento Nervioso/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Restricción Física/efectos adversos
15.
J Neural Transm (Vienna) ; 119(6): 669-77, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22160488

RESUMEN

Calcineurin (CaN)-mediated excitotoxicity impairs γ-aminobutyric acid (GABA) transmission and induces neuronal apoptosis. Ca(2+)-dependent K(+)-Cl(-) cotransporter 2 (KCC2) participates in GABAergic inhibitory transmission. However, the mechanism by which CaN mediates GABA receptor-mediated KCC2 in seizures is not fully understood. In the present study, we investigated the altered expression of KCC2 and the effects of the CaN inhibitor FK506 on KCC2 expression in the mouse hippocampus following kainic acid (KA) treatment. FK506 was injected twice 24 h and 30 min before KA treatment and then mice were treated with KA and killed 2 days later. FK506 had anticonvulsant effect on KA-induced seizure activities. CaN cleavage was evident in the hippocampus 24 h after KA treatment. FK506 pretreatment blocked the truncation of CaN in the KA-treated hippocampus. Cresyl violet and TUNEL staining showed that FK506 prevented KA-induced hippocampal cell death. In particular, Western blot analysis showed that KCC2 expression was time dependent, with a peak at 6 h and a return to decreased levels at 48 h, whereas FK506 pretreatment inhibited the KA-induced decrease in KCC2 expression in the hippocampus. Immunofluorescence showed that FK506 pretreatment protected the loss of inhibitory GABAergic KCC2-expressing neurons following KA treatment. Taken together, these results provide evidence that altered KCC2 expression may be associated with Ca(2+)-mediated seizure activity and indicate that neuron-specific KCC2 may be involved in neuroprotection after seizures.


Asunto(s)
Inhibidores de la Calcineurina , Hipocampo/metabolismo , Inmunosupresores/farmacología , Estado Epiléptico/metabolismo , Simportadores/biosíntesis , Tacrolimus/farmacología , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores , Técnica del Anticuerpo Fluorescente , Hipocampo/efectos de los fármacos , Hipocampo/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ácido Kaínico , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología , Cotransportadores de K Cl
16.
Int J Cancer ; 128(6): 1316-26, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20506295

RESUMEN

Chitinase 3-like 1 (CHI3L1) is a secreted glycoprotein that has pleiotropic activity in aggressive cancers. In our study, we examined the expression and function of CHI3L1 in glioma cells. CHI3L1 was highly expressed in human glioma tissue, whereas its expression in normal brain tissue was very low. CHI3L1 suppression by shRNA reduced glioma cell invasion, anchorage-independent growth and increased cell death triggered by several anticancer drugs, including cisplatin, etoposide and doxorubicin, whereas CHI3L1 overexpression had the opposite effect in glioma cells. Because the invasive nature of glioma cells plays a critical role in the high morbidity of glioma, we have further defined the role of CHI3L1 in the process of glioma invasion. Downregulation of CHI3L1 results in decreased cell-matrix adhesion and causes a marked increase in stress fiber formation and cell size with fewer cellular processes. Furthermore, the expression and activity of matrix metalloproteinase-2 was also decreased in glioma cells in which CHI3L1 was knocked down. Taken together, these results suggest that CHI3L1 plays an important role in the regulation of malignant transformation and local invasiveness in gliomas. Thus, targeting the CHI3L1 molecule may be a potential therapeutic molecular target for gliomas.


Asunto(s)
Apoptosis , Biomarcadores de Tumor/metabolismo , Glioma/metabolismo , Glioma/patología , Glicoproteínas/metabolismo , Lectinas/metabolismo , Adipoquinas , Biomarcadores de Tumor/genética , Northern Blotting , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteína 1 Similar a Quitinasa-3 , Glioma/genética , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/genética , Humanos , Técnicas para Inmunoenzimas , Lectinas/antagonistas & inhibidores , Lectinas/genética , Invasividad Neoplásica , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
17.
Biochem Biophys Res Commun ; 393(3): 476-80, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20152803

RESUMEN

Kainic acid (KA) induces hippocampal cell death and astrocyte proliferation. There are reports that sphingosine kinase (SPHK)1 and sphingosine-1- phosphate (S1P) receptor 1 (S1P(1)) signaling axis controls astrocyte proliferation. Here we examined the temporal changes of SPHK1/S1P(1) in mouse hippocampus during KA-induced hippocampal cell death. Mice were killed at 2, 6, 24, or 48 h after KA (30 mg/kg) injection. There was an increase in Fluoro-Jade B-positive cells in the hippocampus of KA-treated mice with temporal changes of glial fibrillary acidic protein (GFAP) expression. The lowest level of SPHK1 protein expression was found 2h after KA treatment. Six hours after KA treatment, the expression of SPHK1 and S1P(1) proteins steadily increased in the hippocampus. In immunohistochemical analysis, SPHK1 and S1P(1) are more immunoreactive in astrocytes within the hippocampus of KA-treated mice than in hippocampus of control mice. These results indicate that SPHK1/S1P(1) signaling axis may play an important role in astrocytes proliferation during KA-induced excitotoxicity.


Asunto(s)
Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Receptores de Lisoesfingolípidos/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/biosíntesis , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología
18.
Cell Rep ; 30(4): 1063-1076.e5, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31995750

RESUMEN

The resolution phase of acute inflammation is essential for tissue homeostasis, yet the underlying mechanisms remain unclear. We demonstrate that resolution of inflammation involves interactions between CD38 and tristetraprolin (TTP). During the onset of acute inflammation, CD38 levels are increased, leading to the production of Ca2+-signaling messengers, nicotinic acid adenine dinucleotide phosphate (NAADP), ADP ribose (ADPR), and cyclic ADPR (cADPR) from NAD(P)+. To initiate the onset of resolution, TTP expression is increased by the second messengers, NAADP and cADPR, which downregulate CD38 expression. The activation of TTP by Sirt1-dependent deacetylation, in response to increased NAD+ levels, suppresses the acute inflammatory response and decreases Rheb expression, inhibits mTORC1, and induces autophagolysosomes for bacterial clearance. TTP may represent a mechanistic target of anti-inflammatory agents, such as carbon monoxide. TTP mediates crosstalk between acute inflammation and autophagic clearance of bacteria from damaged tissue in the resolution of inflammation during sepsis.


Asunto(s)
ADP-Ribosil Ciclasa 1/inmunología , Inflamación/metabolismo , Glicoproteínas de Membrana/inmunología , Sepsis/metabolismo , Tristetraprolina/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/inmunología , Autofagosomas/metabolismo , Autofagosomas/microbiología , Calcio/metabolismo , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacología , Línea Celular , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NAD/metabolismo , NADP/metabolismo , ARN Interferente Pequeño , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Sepsis/enzimología , Sepsis/inmunología , Sirtuina 1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tristetraprolina/genética
19.
Biochem Biophys Res Commun ; 388(2): 256-60, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19664600

RESUMEN

Transthyretin (TTR) is a human disease-associated amyloidogenic protein that has been implicated in senile systemic amyloidosis (SSA) and familial amyloidotic polyneuropathy (FAP). FAP typically results in severe and early-onset disease, and the only therapy established so far is liver transplantation; thus, developing new strategies for treating FAP is of paramount interest. Clusterin has recently been proposed to play a role as an extracellular molecular chaperone, affecting the fibril formation of amyloidogenic proteins. The ability of clusterin to influence amyloid fibril formation prompted us to investigate whether clusterin is capable of inhibiting TTR amyloidosis. Here, we report that clusterin strongly interacts with wild-type TTR and TTR variants V30M and L55P under acidic conditions, and blocks the amyloid fibril formation of TTR variants. In particular, the amyloid fibril formation of V30M TTR in the presence of clusterin is reduced to level similar to wild-type TTR. We also demonstrated that clusterin is an effective inhibitor of L55P TTR amyloidosis, the most aggressive form of TTR diseases. The mechanism by which clusterin inhibits TTR amyloidosis appears to be through stabilization of TTR tetrameric structure. These findings suggest the possibility of using clusterin as a therapeutic agent for TTR amyloidosis.


Asunto(s)
Neuropatías Amiloides Familiares/metabolismo , Amiloide/antagonistas & inhibidores , Clusterina/metabolismo , Prealbúmina/metabolismo , Amiloide/metabolismo , Neuropatías Amiloides Familiares/tratamiento farmacológico , Animales , Línea Celular Tumoral , Clusterina/uso terapéutico , Humanos , Prealbúmina/genética , Ratas
20.
Crit Care Med ; 37(6): 2033-44, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19384198

RESUMEN

OBJECTIVE: Tubular cell apoptosis is linked to the development of acute kidney injury (AKI), which is a frequent complication of traumatic rhabdomyolysis. The 14-3-3 protein, a multifunctional regulatory protein, binds a variety of apoptotic proteins and is a target of c-Jun N-terminal kinase (JNK) in the cell death signaling pathway. Therefore, we examined whether JNK phosphorylates 14-3-3 and downstream mitochondrial death pathway mediates apoptosis in myoglobinuric acute kidney injury to determine whether these events are regulated by glutamine, which is known to induce heat shock protein 70 (Hsp70), or involved in the synthesis of glutathione (GSH). DESIGN: A prospective, randomized, controlled animal trial. SETTING: University research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: We utilized a rat model of myoglobinuric AKI. Glutamine or saline was administered intraperitoneally before and after glycerol injection. Apoptotic cell death was determined via transferase-mediated deoxyuridine triphosphate nick-end labeling staining, and Hsp70, JNK, phospho-JNK, 14-3-3, phospho-14-3-3, and many other apoptotic proteins were examined via Western blot. Relative interactions between these proteins were tested by coimmunoprecipitation analyses. Also, GSH levels were determined to further test whether glutamine affects apoptotic cell death in myoglobinuric AKI. MEASUREMENTS AND MAIN RESULTS: Glutamine treatment elevated levels of Hsp70 or reduced GSH and attenuated tubular cell apoptosis in kidney tissues of rats with myoglobinuric AKI. Further, Hsp70 physically associated with JNK, thereby limiting its activation. In addition, JNK evidently interacted with 14-3-3, leading to its phosphorylation, Bad or Bax dissociation from 14-3-3, and subsequent Bax mitochondrial translocation and caspase activation in rats with acute renal failure. Glutamine treatment very modestly lowered elevated levels of serum creatinine in AKI rats. CONCLUSIONS: A signaling link between JNK and 14-3-3 and subsequent mitochondrial death pathway may partly act as an early signaling that promotes apoptotic cell death leading to AKI, and glutamine may at least partially prevent apoptosis via enhancing Hsp70 or GSH levels.


Asunto(s)
Proteínas 14-3-3/metabolismo , Lesión Renal Aguda/enzimología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Glutamina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Túbulos Renales Distales/citología , Túbulos Renales Distales/efectos de los fármacos , Lesión Renal Aguda/etiología , Animales , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA