Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Gan To Kagaku Ryoho ; 46(8): 1327-1329, 2019 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-31501381

RESUMEN

BACKGROUND: This study was designed to clarify effects of postoperative adjuvant chemotherapy for high-risk Stage Ⅱ colorectal cancer. METHOD: The subjects were 99 patients with high-risk Stage Ⅱcolorectal cancer who underwent surgery at our department from October 2013 to March 2018. Patients were classified into adjuvant chemotherapy group and nonadjuvant chemotherapy group. Overall survival(OS)and recurrence-free survival(RFS)were analyzed between the 2 groups. RESULTS: Thirty six patients(36.4%)underwent adjuvant chemotherapy. Adjuvant chemotherapy group were younger(p<0.010), had a better ASA-PS(p<0.010), good preoperative Hb(p<0.010), and preoperative Alb(p<0.010)compared to non-adjuvant chemotherapy group. There was no difference between the 2 groups in the high-risk factors for recurrence. Most patient had an oral medication as for adjuvant chemotherapy. There was no difference in OS and RFS between the 2 groups. CONCLUSION: Postoperative adjuvant chemotherapy for high-risk Stage Ⅱ colorectal cancer did not significantly improve the OS and RFS. Further study is necessary to asses the suitable regimen and patients eligible for chemotherapy.


Asunto(s)
Neoplasias del Colon , Protocolos de Quimioterapia Combinada Antineoplásica , Quimioterapia Adyuvante , Neoplasias del Colon/tratamiento farmacológico , Humanos , Recurrencia Local de Neoplasia , Estadificación de Neoplasias
2.
J Neurochem ; 145(6): 474-488, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29500815

RESUMEN

Sepsis-associated encephalopathy (SAE), characterized as diffuse brain dysfunction and neurological manifestations secondary to sepsis, is a common complication in critically ill patients and can give rise to poor outcome, but understanding the molecular basis of this disorder remains a major challenge. Given the emerging role of G protein-coupled receptor 2 (GRK2), first identified as a G protein-coupled receptor (GPCR) regulator, in the regulation of non-G protein-coupled receptor-related molecules contributing to diverse cellular functions and pathology, including inflammation, we tested the hypothesis that GRK2 may be linked to the neuropathogenesis of SAE. When mouse MG6 microglial cells were challenged with lipopolysaccharide (LPS), GRK2 cytosolic expression was highly up-regulated. The ablation of GRK2 by small interfering RNAs (siRNAs) prevented an increase in intracellular reactive oxygen species generation in LPS-stimulated MG6 cells. Furthermore, the LPS-induced up-regulation of inducible nitric-oxide synthase expression and increase in nitric oxide production were negated by GRK2 inhibitor or siRNAs. However, GRK2 inhibition was without effect on overproduction of tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß in LPS-stimulated MG cells. In mice with cecal ligation and puncture-induced sepsis, treatment with GRK2 inhibitor reduced high levels of oxidative and nitrosative stress in the mice brains, where GRK2 expression was up-regulated, alleviated neurohistological damage observed in cerebral cortex sections, and conferred a significant survival advantage to CLP mice. Altogether, these results uncover the novel role for GRK2 in regulating cellular oxidative and nitrosative stress during inflammation and suggest that GRK2 may have a potential as an intriguing therapeutic target to prevent or treat SAE.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Encefalopatía Asociada a la Sepsis/patología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Citocinas/biosíntesis , Inhibidores Enzimáticos/uso terapéutico , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Interferente Pequeño/farmacología , Encefalopatía Asociada a la Sepsis/complicaciones , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
3.
Crit Care Med ; 43(11): e508-20, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26468714

RESUMEN

OBJECTIVES: The calcium sensitizer levosimendan is used in treatment of decompensated heart failure and may also exhibit anti-inflammatory properties. We examined whether treatment with levosimendan is substantially beneficial in mice with cecal ligation and puncture-induced polymicrobial sepsis, and its arbitration mechanism was explored in the mouse macrophage cell line RAW264.7. DESIGN: Laboratory and animal/cell research. SETTING: University research laboratory. SUBJECTS: BALB/c mice (8-10 wk old) and mouse macrophage cell line RAW264.7 cells. INTERVENTIONS: Levosimendan (0.5 µg/kg/min) was administered to mice through an osmotic pump that was implanted into the peritoneal cavity immediately following surgery. In RAW264.7 cells, levosimendan was added to the culture medium 30 minutes before lipopolysaccharide. MEASUREMENTS AND MAIN RESULTS: When levosimendan was continuously administered to cecal ligation and puncture-induced septic mice, a significant improvement of left ventricular function was found without any change in heart rate, and hypotension was significantly mitigated. Furthermore, levosimendan conferred substantial protection against sepsis-associated inflammation in mice, as indicated by reduced lung injury and decreased blood proinflammatory and chemotactic cytokine levels. These beneficial effects of levosimendan led to a significant improvement of survival in mice after cecal ligation and puncture. In endotoxin-stimulated RAW264.7 macrophages, treatment with levosimendan and pimobendan suppressed overproduction of proinflammatory and chemotactic cytokines. Levosimendan and pimobendan were without effect on activation of the nuclear factor-κB, mitogen-activated protein kinase, and Akt pathways. Instead, levosimendan and pimobendan prevented high mobility group box 1 release from the nucleus to the extracellular space in macrophages. This was associated with inhibition of the Rho kinase signaling pathway. The elevated serum high mobility group box 1 levels in cecal ligation and puncture-induced septic mice were also inhibited by continued administration of levosimendan and pimobendan. CONCLUSIONS: We define a novel mechanism for the anti-inflammatory action of levosimendan and suggest that the pharmacological profiles of levosimendan as both an inotrope and an anti-inflammatory agent could contribute to its clinical benefit in patients with sepsis with heart problems.


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/metabolismo , Hidrazonas/farmacología , Macrófagos/efectos de los fármacos , Piridazinas/farmacología , Sepsis/tratamiento farmacológico , Sepsis/patología , Animales , Biopsia con Aguja , Western Blotting , Ciego/cirugía , Células Cultivadas , Citocinas/efectos de los fármacos , Modelos Animales de Enfermedad , Ecocardiografía/métodos , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Inyecciones Intravenosas , Ligadura/métodos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Valores de Referencia , Sepsis/mortalidad , Simendán , Estadísticas no Paramétricas , Tasa de Supervivencia
4.
J Surg Res ; 187(2): 559-70, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24290430

RESUMEN

BACKGROUND: Epigenetic programming, dynamically regulated by histone acetylation, may play a key role in the pathophysiology of sepsis. We examined whether histone deacetylase (HDAC) can contribute to sepsis-associated inflammation and apoptosis. MATERIALS AND METHODS: Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in BALB/c mice. An intraperitoneal injection of CG200745 (10 mg/kg), a novel broad-spectrum HDAC inhibitor, or valproic acid (500 mg/kg), a predominant inhibitor of class I HDACs, was given 3 h before surgery. RESULTS: HDAC1, HDAC2, and HDAC3 protein levels were decreased in lungs after CLP. Furthermore, CLP-induced sepsis increased both histone H3 and H4 acetylation levels in lungs. When CG200745 was given, apoptosis induction was strongly suppressed in lungs and spleens of septic mice. This antiapoptotic effect of CG200745 was not accompanied by upregulation of antiapoptotic and downregulation of proapoptotic Bcl-2 family member proteins. Treatment with CG200745 failed to inhibit elevated levels of serum cytokines and prevent lung inflammation in septic mice. Valproic acid also showed antiapoptotic but not anti-inflammatory effects in septic mice. CONCLUSIONS: These findings imply that HDAC inhibitors are a unique agent to prevent cell apoptosis in sepsis at their doses that do not improve inflammatory features, indicating that septic inflammation and apoptosis may not necessarily be essential for one another's existence. This study also represents the first report that CLP-induced sepsis downregulates HDACs. Nevertheless, the data with HDAC inhibitors suggest that imbalance in histone acetylation may play a contributory role in expression or repression of genes involved in septic cell apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Naftalenos/farmacología , Neumonía/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Animales , Epigenómica , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/metabolismo , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Neumonía/metabolismo , Neumonía/patología , Sepsis/metabolismo , Sepsis/patología , Bazo/enzimología , Bazo/patología
5.
Chemosphere ; 313: 137526, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36513194

RESUMEN

Biogenic manganese (Mn) oxides occur ubiquitously in the environment including the uranium (U) mill tailings at the Ningyo-toge U mine in Okayama, Japan, being important in the sequestration of radioactive radium. To understand the nanoscale processes in Mn oxides formation at the U mill tailings site, Mn2+ absorption by a basidiomycete fungus, Coprinopsis urticicola, isolated from Ningyo-toge mine water samples, was investigated in the laboratory under controlled conditions utilizing electron microscopy, synchrotron-based X-ray analysis, and fluorescence microscopy with a molecular pH probe. The fungus' growth was first investigated in an agar-solidified medium supplemented with 1.0 mmol/L Mn2+, and Cu2+ (0-200 µM), Zn2+ (0-200 µM), or diphenyleneiodonium (DPI) chloride (0-100 µM) at 25 °C. The results revealed that Zn2+ has no significant effects on Mn oxide formation, whereas Cu2+ and DPI significantly inhibit both fungal growth and Mn oxidation, indicating superoxide-mediated Mn oxidation. Indeed, nitroblue tetrazolium and diaminobenzidine assays on the growing fungus revealed the production of superoxide and peroxide. During the interaction of Mn2+ with the fungus in solution medium at the initial pH of 5.67, a small fraction of Mn2+ infiltrated the fungal hyphae within 8 h, forming a few tens of nm-sized concentrates of soluble Mn2+ in the intracellular pH of ∼6.5. After 1 day of incubation, Mn oxides began to precipitate on the hyphae, which were characterized as fibrous nanocrystals with a hexagonal birnessite-structure, these forming spherical aggregates with a diameter of ∼1.5 µm. These nanoscale processes associated with the fungal species derived from the Ningyo-toge mine area provide additional insights into the existing mechanisms of Mn oxidation by filamentous fungi at other U mill tailings sites under circumneutral pH conditions. Such processes add to the class of reactions important to the sequestration of toxic elements.


Asunto(s)
Basidiomycota , Superóxidos , Óxidos/química , Compuestos de Manganeso/química , Oxidación-Reducción , Hongos
6.
Am J Physiol Lung Cell Mol Physiol ; 303(2): L130-40, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22610350

RESUMEN

Olprinone, a specific phosphodiesterase III inhibitor, and corforsin daropate, a direct adenylate cyclase activator, are now being used in critical conditions. We investigated whether their therapeutic use provides protection against septic acute lung injury (ALI) and mortality. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in BALB/c mice. Olprinone or colforsin daropate was continuously given through an osmotic pump that was implanted into the peritoneal cavity immediately following CLP. These treatments prevented the ALI development in CLP mice, as indicated by the findings that severe hypoxemia, increased pulmonary vascular permeability, and histological lung damage were strikingly remedied. Furthermore, continued administration of olprinone or colforsin daropate suppressed apoptosis induction in septic lungs and improved the survival of CLP mice. Olprinone and corforsin daropate enhanced Akt phosphorylation in septic lungs. Wortmannin, which inhibits the Akt upstream regulator phosphatidylinositol 3-kinase, abrogated the protective effects of olprinone and corforsin daropate on sepsis-associated lung inflammation and apoptosis. In vivo transfection of cyclic AMP response element binding protein (CREB) decoy oligodeoxynucleotide failed to negate the abilities of these agents to increase Akt phosphorylation and to inhibit IκBα degradation in septic lungs. These results demonstrate for the first time that CREB-independent Akt-mediated signaling is a critical mechanism contributing to the therapeutic effects of olprinone and corforsin daropate on septic ALI. Moreover, our data also suggest that these cyclic AMP-related agents, by blocking both nuclear factor-κB activation and apoptosis induction, may represent an effective therapeutic approach to the treatment of the septic syndrome.


Asunto(s)
Apoptosis/efectos de los fármacos , Colforsina/análogos & derivados , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Imidazoles/farmacología , Neumonía/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridonas/farmacología , Choque Séptico/tratamiento farmacológico , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/patología , Androstadienos/farmacología , Animales , Ciego/microbiología , Ciego/patología , Colforsina/farmacología , Colforsina/uso terapéutico , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Activación Enzimática , Activadores de Enzimas/farmacología , Activadores de Enzimas/uso terapéutico , Hipotensión/microbiología , Imidazoles/uso terapéutico , Ligadura , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neumonía/microbiología , Neumonía/patología , Piridonas/uso terapéutico , Choque Séptico/sangre , Choque Séptico/microbiología , Transducción de Señal , Wortmanina
7.
J Pharmacol Exp Ther ; 336(2): 381-90, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21057058

RESUMEN

There is growing evidence that the HMG-CoA reductase inhibitors (statins) provide some of the beneficial effects that are independent of their lipid-lowering effects. Recent animal experiments and clinical trials suggest that statin use may limit the development of sepsis and associated systemic inflammation. The aim of this study was to explore the potential role of statins in the prevention treatment of sepsis-induced acute lung injury (ALI). Mice were rendered septic by cecal ligation and puncture (CLP). An intraperitoneal injection of 3 mg/kg per day of pitavastatin was initiated 4 days before surgery and was maintained for life support afterward, which significantly improved the survival of CLP mice. Treatment with pitavastatin prevented the ALI development in CLP mice, as indicated by the findings that severe hypoxemia, increased pulmonary vascular permeability, and histological lung damage, including inflammatory cell infiltrate, were greatly remedied. This was associated with down-regulation of increased activity of nuclear factor-κB (NF-κB) in septic lungs. Although plasma cortisol showed a sharp rise, glucocorticoid receptor (GCR) expression in the lungs was strikingly reduced after the onset of CLP-induced sepsis. It is noteworthy that pitavastatin increased GCR expression with an increase in alveolar macrophages in which GCRs are localized, without modifying the sepsis-associated rise in plasma cortisol. These results confirm significant protection by pitavastatin on septic ALI and demonstrate that down-regulated NF-κB activation associated with the GCR expression increase consequent to the increased number of alveolar macrophages may explain, in part, the mechanisms responsible for favorable effects of statins on the ALI management.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Macrófagos Alveolares/fisiología , Quinolinas/uso terapéutico , Receptores de Glucocorticoides/fisiología , Sepsis/tratamiento farmacológico , Animales , Apoptosis , Citocinas/biosíntesis , Hidrocortisona/sangre , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Sepsis/complicaciones , Sepsis/patología
8.
Cardiovasc Diabetol ; 10: 73, 2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21816064

RESUMEN

BACKGROUND: High glucose can induce apoptosis in vascular endothelial cells, which may contribute to the development of vascular complications in diabetes. We evaluated the role of the death receptor pathway of apoptotic signaling in high glucose-induced apoptosis in human coronary artery endothelial cells (HCAECs). METHODS: HCAECs were treated with media containing 5.6, 11.1, and 16.7 mM of glucose for 24 h in the presence or absence of tumor necrosis factor (TNF)-α. For detection of apoptosis, DNA fragmentation assay was used. HCAEC expression of death receptors were analyzed by the PCR and flow cytometry methods. Also, using immunohistochemical techniques, coronary expression of death receptors was assessed in streptozotocin-nicotinamide-induced type 2 diabetic mice. RESULTS: Exposure of HCAECs to high glucose resulted in a significant increase in TNF-R1 and Fas expression, compared with normal glucose. High glucose increased TNF-α production by HCAECs and exogenous TNF-α up-regulated TNF-R1 and Fas expression in HCAECs. High glucose-induced up-regulation of TNF-R1 and Fas expression was undetectable in the presence of TNF-α. Treatment with TNF-R1 neutralizing peptides significantly inhibited high glucose-induced endothelial cell apoptosis. Type 2 diabetic mice displayed appreciable expression of TNF-R1 and Fas in coronary vessels. CONCLUSIONS: In association with increased TNF-α levels, the death receptors, TNF-R1 and Fas, are up-regulated in HCAECs under high glucose conditions, which could in turn play a role in high glucose-induced endothelial cell apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Glucosa/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Apoptosis/fisiología , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estreptozocina/efectos adversos , Regulación hacia Arriba/fisiología
9.
J Pharmacol Exp Ther ; 332(3): 730-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20008488

RESUMEN

The histamine H(4) receptor is the most recently identified receptor and is considered to play a role in a variety of inflammatory diseases. Histamine levels in the plasma are known to be elevated in animal models of sepsis and in septic patients. The aim of this study was to test the hypothesis that the H(4) receptor may play a significant role in the pathophysiology of sepsis. Polymicrobial sepsis was induced by cecal ligation and puncture in BALB/c mice. Although the H(4) receptor gene was undetectable in normal peripheral key organs, with the exception of the spleen, the expression levels of this gene were highly up-regulated in all those organs of septic mice. In vivo transfection of nuclear factor-kappaB (NF-kappaB) decoy oligodeoxynucleotide, but not of its scrambled form, resulted in a great inhibition of sepsis-induced overexpression of the H(4) receptor gene. In septic mice, marked increases in caspase-3 activation and follicular lymphocyte apoptosis in spleens were strongly suppressed by systemic treatment with synthetic small interfering RNA (siRNA) targeted to the H(4) receptor. This was associated with the up-regulation of a number of antiapoptotic proteins. These antiapoptotic effects of H(4) receptor siRNA treatment were all inhibited by further application of NF-kappaB decoy oligonucleotide. Our results suggest that superinduction of the histamine H(4) receptor gene in peripheral key organs, including the spleen, that is promoted by sepsis is transcriptionally controlled by NF-kappaB, whereas stimulation of this receptor is involved in the development of sepsis-induced splenic apoptosis through counteraction of the antiapoptotic action of NF-kappaB.


Asunto(s)
Apoptosis , FN-kappa B/fisiología , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Histamínicos/biosíntesis , Sepsis/metabolismo , Sepsis/patología , Bazo/patología , Animales , Caspasa 3/metabolismo , Activación Enzimática , Linfocitos/patología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , Oligonucleótidos/farmacología , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos/genética , Receptores Histamínicos H4 , Bazo/metabolismo , Transcripción Genética , Regulación hacia Arriba
10.
J Pharmacol Sci ; 114(4): 354-65, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21081836

RESUMEN

Sepsis remains the leading cause of death in critically ill patients. A major problem contributing to sepsis-related high mortality is the lack of effective medical treatment. Thus, the key goal in critical care medicine is to develop novel therapeutic strategies that will impact favorably on septic patient outcome. While it is generally accepted that sepsis is an inflammatory state resulting from the systemic response to infection, apoptosis is implicated to be an important mechanism of the death of lymphocytes, gastrointestinal and lung epithelial cells, and vascular endothelial cells associated with the development of multiple organ failure in sepsis. The pivotal role of cell apoptosis is now highlighted by multiple studies demonstrating that prevention of cell apoptosis can improve survival in clinically relevant animal models of sepsis. In this review article, we address the scientific rationale for remedying apoptotic cell death in sepsis and propose that therapeutic efforts aimed at blocking cell signaling pathways leading to apoptosis may represent an attractive target for sepsis therapy.


Asunto(s)
Apoptosis/fisiología , Terapia Molecular Dirigida , Sepsis/etiología , Sepsis/terapia , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/genética , Caspasas/fisiología , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Células Epiteliales/patología , Silenciador del Gen , Glucocorticoides/administración & dosificación , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , Humanos , Linfocitos/patología , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal/efectos de los fármacos
11.
Sci Rep ; 10(1): 15316, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943679

RESUMEN

Sepsis is a major clinical challenge with unacceptably high mortality. The signal transducers and activators of transcription (STAT) family of transcription factors is known to activate critical mediators of cytokine responses, and, among this family, STAT3 is implicated to be a key transcription factor in both immunity and inflammatory pathways. We investigated whether in vivo introduction of synthetic double-stranded STAT3 decoy oligodeoxynucleotides (ODNs) can provide benefits for reducing organ injury and mortality in mice with cecal ligation and puncture (CLP)-induced polymicrobial sepsis. We found that STAT3 was rapidly activated in major end-organ tissues following CLP, which was accompanied by activation of the upstream kinase JAK2. Transfection of STAT3 decoy ODNs downregulated pro-inflammatory cytokine/chemokine overproduction in CLP mice. Moreover, STAT3 decoy ODN transfection significantly reduced the increases in tissue mRNAs and proteins of high mobility group box 1 (HMGB1) and strongly suppressed the excessive elevation in serum HMGB1 levels in CLP mice. Finally, STAT3 decoy ODN administration minimized the development of sepsis-driven major end-organ injury and led to a significant survival advantage in mice after CLP. Our results suggest a critical role of STAT3 in the sepsis pathophysiology and the potential usefulness of STAT3 decoy ODNs for sepsis gene therapy.


Asunto(s)
Ciego/metabolismo , Oligodesoxirribonucleótidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Sepsis/metabolismo , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Janus Quinasa 2/metabolismo , Ligadura/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Punciones/métodos , ARN Mensajero/metabolismo , Transfección/métodos
12.
Pharmacol Ther ; 120(3): 333-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18930078

RESUMEN

Cellular senescence is characterized by permanent exit from the cell cycle and the appearance of distinct morphological and functional changes associated with an impairment of cellular homeostasis. Many studies support the occurrence of vascular endothelial cell senescence in vivo, and the senescent phenotype of endothelial cells can be transformed from anti-atherosclerotic to pro-atherosclerotic. Thus, endothelial cell senescence promotes endothelial dysfunction and may contribute to the pathogenesis of age-associated vascular disorders. Emerging evidence suggests that increasing nitric oxide (NO) bioavailability or endothelial NO synthase (eNOS) activity activates telomerase and delays endothelial cell senescence. In this review, we discuss the potential mechanisms underlying the ability of NO to prevent endothelial cell senescence and describe the possible changes in the NO-mediated anti-senescence effect under pathophysiological conditions, including oxidative stress and hyperglycemia. Further understanding of the mechanisms underlying the anti-senescence effect of NO in endothelial cells will provide insights into the potential of eNOS-based anti-senescence therapy for age-associated vascular disorders.


Asunto(s)
Senescencia Celular/fisiología , Endotelio Vascular/fisiología , Óxido Nítrico/fisiología , Animales , Endotelio Vascular/metabolismo , Estrógenos/fisiología , Glucosa/metabolismo , Humanos , Telómero/fisiología
13.
Crit Care Med ; 37(10): 2791-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19707125

RESUMEN

OBJECTIVE: Insulin-resistant hyperglycemia is commonly observed in septic patients and may actually lead to some of adverse outcomes. We examined the changes in insulin signaling and glucose uptake regulation in sepsis and the involvement of the nuclear factor-kappaB pathway. DESIGN: Controlled animal study. SETTING: University research laboratory. SUBJECTS: One hundred fifty-four BALB/c mice (8-12 wks of age). INTERVENTIONS: The following four experimental groups were studied: sham-operated control, cecal ligation and puncture-induced sepsis, sepsis + nuclear factor-kappaB decoy oligodeoxynucleotide treatment, and sepsis + scrambled decoy oligodeoxynucleotide treatment. MEASUREMENTS AND MAIN RESULTS: Septic mice were markedly hyperinsulinemic with apparently normal blood glucose levels in the fasted state, suggesting they are insulin-resistant. In fact, glucose clearance in response to insulin was markedly impaired in septic mice. They had impaired GLUT4 membrane translocation resulting from impaired insulin signaling as indicated by the decreased amount of insulin receptor substrate protein and the reduced activation of phosphatidylinositol 3-kinase and Akt. Interestingly, injection of nuclear factor-kappaB decoy oligodeoxynucleotide into the skeletal muscle dramatically improved all of the changes, including glucose clearance and insulin signaling. We also found that the Cbl-associated protein to TC10 pathway, another pathway regulating GLUT4 translocation, was up-regulated in septic mice in a nuclear factor-kappaB-dependent manner. This pathway may be one of the compensatory mechanisms to translocate GLUT4 because silencing of the individual components of the pathway with small interfering RNAs further reduced GLUT4 translocation in muscles of septic mice. CONCLUSIONS: In sepsis, skeletal muscle GLUT4 translocation is impaired as a result of the reduced phosphatidylinositol 3-kinase/Akt pathway associated with insulin receptor substrate down-regulation through nuclear factor-kappaB activation.


Asunto(s)
Glucemia/metabolismo , Transportador de Glucosa de Tipo 4/genética , Resistencia a la Insulina/genética , Oligodesoxirribonucleótidos/genética , Sepsis/genética , Transfección , Animales , Modelos Animales de Enfermedad , Hiperinsulinismo/genética , Perforación Intestinal/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/metabolismo , Proteína Oncogénica v-akt/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Translocación Genética
14.
Naunyn Schmiedebergs Arch Pharmacol ; 391(9): 1021-1032, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29922941

RESUMEN

Levosimendan and milrinone may be used in place of dobutamine to increase cardiac output in septic patients with a low cardiac output due to impaired cardiac function. The effects of the two inotropic agents on cardiac inflammation and left ventricular (LV) performance were examined in mice with cecal ligation and puncture (CLP)-induced sepsis. CLP mice displayed significant cardiac inflammation, as indicated by highly increased pro-inflammatory cytokines and neutrophil infiltration in myocardial tissues. When continuously given, levosimendan prevented but milrinone exaggerated cardiac inflammation, but they significantly reduced the elevations in plasma cardiac troponin-I and heart-type fatty acid-binding protein, clinical markers of cardiac injury. Echocardiographic assessment of cardiac function showed that the effect of levosimendan, given by an intravenous bolus injection, on LV performance was impaired in CLP mice, whereas milrinone produced inotropic responses equally in sham-operated and CLP mice. A lesser effect of levosimendan on LV performance after CLP was also found in spontaneously beating Langendorff-perfused hearts. In ventricular myocytes isolated from control and CLP mice, levosimendan, but not milrinone, caused a large increase in the L-type calcium current. This study represents that levosimendan and milrinone have cardioprotective properties but provide different advantages and drawbacks to cardiac inflammation/dysfunction in sepsis.


Asunto(s)
Cardiotónicos/uso terapéutico , Milrinona/uso terapéutico , Sepsis/tratamiento farmacológico , Simendán/uso terapéutico , Animales , Calcio/fisiología , Ciego/cirugía , Citocinas/genética , Ligadura , Masculino , Ratones Endogámicos BALB C , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Sepsis/etiología , Sepsis/genética , Sepsis/fisiopatología , Función Ventricular Izquierda/efectos de los fármacos , Heridas Penetrantes/complicaciones
15.
J Neurochem ; 103(5): 1883-96, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17883398

RESUMEN

In cultured bovine adrenal chromaffin cells, 48 h-treatment with 20 mmol/L LiCl, 1 mmol/L valproic acid, 30 micromol/L SB216763, 30 micromol/L SB415286, or 100 nmol/L insulin, a condition that inhibits constitutive active glycogen synthase kinase-3 (GSK-3), decreased cell surface (125)I-insulin binding capacity by approximately 39%, without altering the K(d) value; LiCl, SB216763 or insulin decreased insulin receptor (IR) and IR precursor levels, attenuating insulin-induced Tyr-autophosphorylation of IR. LiCl increased inhibitory Ser9-phosphorylation of GSK-3beta at 6 h, decreasing (125)I-insulin binding at 24 h. SB216763-induced (125)I-insulin binding reduction (IC(50) = 3 micromol/L) was preceded by beta-catenin level increase by SB216763 (EC(50) = 11 micromol/L), a hallmark of GSK-3 inhibition. Insulin-induced rapid (> 1 min) Ser9-phosphorylation of GSK-3beta (Nemoto et al. 2006) was followed by approximately 48% decrease of IR level. LiCl did not stimulate endocytosis, nor proteolysis of IR. LiCl destabilized IR mRNA (t(1/2) = 9.3 vs. 6.5 h), decreasing IR mRNA level by approximately 47%, without altering IR gene transcription. Decreases of (125)I-insulin binding and IR level, as well as increased Ser9-phosphorylation of GSK-3beta were restored to the control levels by washing the test compound-treated cells. Thus, GSK-3beta regulates IR level via controlling IR mRNA stability.


Asunto(s)
Glándulas Suprarrenales/citología , Células Cromafines/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Estabilidad del ARN/fisiología , Receptor de Insulina/fisiología , Análisis de Varianza , Animales , Bovinos , Células Cultivadas , Células Cromafines/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3 beta , Insulina/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , Ensayo de Unión Radioligante/métodos , Tirosina/metabolismo , beta Catenina/metabolismo
16.
Brain Res ; 1123(1): 12-9, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17074307

RESUMEN

Water homeostasis in the brain is essential for brain function. We have studied how aquaporin (AQP) 1 expression in GP8 immortalized rat brain microvascular endothelial cells is regulated by glucocorticoid. AQP1 protein level was raised by dexamethasone treatment in a time- and concentration-dependent manner. The up-regulation of AQP1 protein by dexamethasone was associated with an increase of AQP1 mRNA level, with no change in the degradation rate of AQP1 mRNA. AQP1 was concentrated in detergent-insoluble fractions in the cells treated with or without dexamethasone, suggesting that function/trafficking of AQP1 may be regulated via the interaction with lipid rafts. Since glucocorticoid therapy has well known beneficial effects in the treatment of brain edema, the induction of AQP1 by dexamethasone raises a possibility that AQP1 plays a role in ameliorating brain edema.


Asunto(s)
Acuaporina 1/metabolismo , Dexametasona/farmacología , Células Endoteliales/metabolismo , Glucocorticoides/farmacología , Microdominios de Membrana/metabolismo , Animales , Acuaporina 1/genética , Encéfalo/irrigación sanguínea , Encéfalo/citología , Capilares/citología , Capilares/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Masculino , Microdominios de Membrana/efectos de los fármacos , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Regulación hacia Arriba
17.
Brain Res ; 1110(1): 1-12, 2006 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16870161

RESUMEN

In cultured bovine adrenal chromaffin cells, 12-h treatment with 1-20 mM LiCl, an inhibitor of glycogen synthase kinase-3 (GSK-3), increased Ser(9) phosphorylation of GSK-3beta by approximately 44%, while decreasing insulin receptor substrate-1 (IRS-1) and IRS-2 protein levels by approximately 38 and approximately 62% in a concentration-dependent manner. Treatment with SB216763 (0.1-30 microM for 12 h), a selective inhibitor of GSK-3, lowered IRS-1 and IRS-2 levels by approximately 38 and approximately 48%, while increasing beta-catenin protein level by approximately 47%, due to the prevention of GSK-3-induced degradation of beta-catenin by SB216763. Insulin (100 nM for 24 h) increased Ser(9) phosphorylation of GSK-3beta by approximately 104%, while decreasing IRS-1 and IRS-2 levels by approximately 41 and approximately 72%; the insulin-induced Ser(9) phosphorylation of GSK-3beta, as well as down-regulations of IRS-1 and IRS-2 levels were restored to the control levels of nontreated cells at 24 h after the washout of the insulin (100 nM for 12 h)-treated cells. Either clasto-lactacystin beta-lactone or lactacystin (an inhibitor of proteasome) prevented LiCl- or SB216763-induced decreases of IRS-1 and IRS-2 levels by approximately 100 and approximately 69%, respectively. In contrast, calpastatin (an inhibitor of calpain) and leupeptin (an inhibitor of lysosome) failed to prevent the decreases of IRS-1 and IRS-2 levels caused by LiCl or SB216763. LiCl or SB216763 lowered IRS-2 mRNA level, with no effect on IRS-1 mRNA level. These results suggest that constitutive activity of GSK-3beta in quiescent cells positively maintains steady-state levels of IRS-1 and IRS-2 via regulating proteasomal degradation and/or synthesis of IRS-1 and IRS-2 proteins.


Asunto(s)
Células Cromafines/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/fisiología , Cloruro de Litio/farmacología , Fosfoproteínas/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Adyuvantes Inmunológicos/farmacología , Glándulas Suprarrenales/citología , Animales , Northern Blotting/métodos , Bovinos , Células Cultivadas , Células Cromafines/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Indoles/farmacología , Lactonas/farmacología , Maleimidas/farmacología , Fosfoproteínas/clasificación , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/farmacología , Serina/metabolismo , Factores de Tiempo
19.
Vascul Pharmacol ; 70: 55-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25869503

RESUMEN

Vascular endothelial cells can absorb higher radiation doses than any other tissue in the body, and post-radiation impaired endothelial nitric oxide synthase (eNOS) function may be developed as a potential contributor to the pathogenesis of vascular injury. In this study, we investigated early alterations of eNOS signaling in human umbilical venous endothelial cells (HUVECs) exposed to X-ray radiation. We found that ionizing radiation increased eNOS phosphorylation at Ser-1177 and dephosphorylation at Thr-495 in HUVECs in a dose-dependent (≤ 20 Gy) and time-dependent (6-72 h) manner. The total expression levels of eNOS were unchanged by radiation. Although a transient but significant increase in extracellular signal-regulated protein kinase 1/2 (ERK1/2) phosphorylation and a biphasic decline in Akt phosphorylation were observed after irradiation, these inhibitors were without effect on the radiation-induced changes in eNOS phosphorylation. There was an increase in protein kinase C-ßII (PKC-ßII) expression and the ablation of PKC-ßII by small interfering RNA (siRNA) negated the radiation effect on the two eNOS phosphorylation events. Furthermore, when the radiation-induced increase in reactive oxygen species (ROS) generation was prevented by the anti-oxidant N-acetyl-L-cysteine, eNOS Ser-1177 phosphorylation and Thr-495 dephosphorylation in irradiated HUVECs were significantly reduced. However, transfection of PKC-ß siRNA did not alter ROS production after irradiation, and NAC failed to block the radiation-induced increase in PKC-ßII expression. Taken together, our results suggest that ionizing radiation-induced eNOS activation in human vascular endothelial cells is attributed to both the up-regulation of PKC-ßII and the increase in ROS generation which were independent of each other.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de la radiación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Quinasa C beta/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/farmacología , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Fosforilación , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C beta/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Regulación hacia Arriba
20.
Front Biosci ; 9: 1954-66, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14977601

RESUMEN

Regulated expression of Na+ channels is indispensable to physiological events, whereas dysregulated expression of otherwise silent or even normal Na+ channel isoforms causes Na+ channelopathies; however, the regulatory mechanisms remain unknown. In quiescent cultured bovine adrenal chromaffin cells, constitutive phosphorylation/activation of extracellular signal-regulated kinase-1 (ERK1) and ERK2 destabilized Nav l.7 Na+ channel alpha-subunit mRNA and decreased its level without altering alpha-subunit gene transcription, thus negatively regulating steady-state level of Na+ channels. Activation of protein kinase C (PKC) down-regulated Na+ channels via PKC isoform-specific mechanisms; conventional PKC-alpha promoted endocytic internalization of Na+ channels, whereas novel PKC-epsilon destabilized alpha-subunit mRNA without altering its gene transcription. Long-lasting (but not short-term) increase of cytoplasmic Ca2+ down-regulated Na+ channels; a slowly-developing moderate increase of Ca2+ activated PKC-alpha and calpain, promoting internalization of Na+ channels, whereas an immediate monophasic and salient plateau increase of Ca2+ lowered alpha- and beta1-subunit mRNA levels. Calcineurin, or FK506 binding protein- and rapamycin-associated protein (FRAP), a serine/threonine protein kinase, down-regulated, whereas insulin receptor tyrosine kinase or protein kinase A (PKA) up-regulated, Na+ channels via modulating Na+ channel internalization, and/or Na+ channel externalization from the trans-Golgi network. Neuroprotective, antiepiletic, antipsychotic, and local anesthetic drugs up-regulated Na+ channels via transcriptional/translational events.


Asunto(s)
Médula Suprarrenal/citología , Células Cromafines/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo , Animales , Calcineurina/fisiología , Señalización del Calcio , Bovinos , Células Cultivadas , Células Cromafines/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fármacos Neuroprotectores/farmacología , Proteína Quinasa C/metabolismo , Proteínas Quinasas/fisiología , Transporte de Proteínas , Receptor de Insulina/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA