Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Physiol ; 233(4): 3638-3647, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29030979

RESUMEN

Pioglitazone is an anti-diabetic agent that belongs to the thiazolidinedione class, which target peroxisome proliferator-activated receptor γ (PPARγ), a transcription factor in the nuclear receptor family. Different cancer cells expressing high levels of PPARγ and PPARγ ligands induce cell cycle arrest, cell differentiation, and apoptosis. However, the mechanisms underlying these processes remain unknown. Here, we investigated the mechanism underlying pioglitazone-induced apoptosis in human cancer cells. We showed that at similar concentrations, pioglitazone induced death in cancer cells expressing high or low levels of PPARγ. Combined treatment of pioglitazone and GW9662, a PPARγ antagonist, did not rescue this cell death phenotype. Z-VAD-fmk, a pan-caspase inhibitor, did not reverse pioglitazone-induced apoptosis in cancer cells expressing PPARγ at high or low levels. Pioglitazone suppressed the activation of signal transducers and activator of transcription 3 (STAT3) and Survivin expression, and enhanced the apoptosis-inducing factor (AIF) levels in these cells. Furthermore, pioglitazone enhanced the cytotoxic effect of cisplatin and oxaliplatin by suppressing Survivin and increasing AIF expression. These results indicated that pioglitazone induced apoptosis via a PPARγ-independent pathway, thus describing pioglitazone as a potential therapeutic agent for controlling the progression of different cancers.


Asunto(s)
PPAR gamma/efectos de los fármacos , Pioglitazona/farmacología , Factor de Transcripción STAT3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/metabolismo , Línea Celular Tumoral , Humanos , Hipoglucemiantes/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , PPAR gamma/metabolismo , Factor de Transcripción STAT3/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factores de Transcripción/metabolismo
2.
J Cell Physiol ; 233(5): 4258-4271, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29057477

RESUMEN

Several autocrine soluble factors, including macrophage inflammatory protein-1α (MIP-1α), tumor necrosis factor-α, and hepatocyte growth factor, promote cell survival and growth in multiple myeloma (MM) cells. We hypothesized that inhibition of the MIP-1α autocrine loop may enhance the cytotoxic effect of anticancer drugs in MM cell lines. In the present study, an MIP-1α neutralizing antibody suppressed cell proliferation and enhanced the cytotoxic effect of melphalan or bortezomib on MM cells. In addition, melphalan resistance cells (RPMI8226/L-PAM and HS-sultan/L-PAM cells) secreted MIP-1α and neutralizing antibody of MIP-1α partially overcame melphalan resistance. Moreover, combination treatment with MIP-1α neutralizing antibody and melphalan or bortezomib inhibited extracellular signal regulated kinase 1/2 (ERK1/2), Akt, and mammalian target of rapamycin (mTOR) activation, Bcl-2, Bcl-xL, and Survivin expression, and upregulated the expression of Bim and cleaved Poly (ADP-ribose) polymerase (PARP). Treatment of IM9 cells with MIP-1α siRNA suppressed the activation of ERK1/2, Akt, and mTOR, and enhanced the cytotoxic effect of melphalan and bortezomib. These results indicate that MIP-1α neutralizing antibodies or MIP-1α siRNA enhance the cytotoxic effect of melphalan and bortezomib by suppressing the chemokine receptor/ERK and chemokine receptor/Akt/mTOR pathways. The inhibition of MIP-1α may thus provide a new therapeutic approach to control tumor progression and bone destruction in patients with MM.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quimiocina CCL3/genética , Resistencia a Antineoplásicos/genética , Mieloma Múltiple/tratamiento farmacológico , Bortezomib/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melfalán/farmacología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Survivin/genética , Serina-Treonina Quinasas TOR/genética , Factor de Necrosis Tumoral alfa/genética , Proteína bcl-X/genética
3.
Exp Physiol ; 102(11): 1435-1447, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28841247

RESUMEN

NEW FINDINGS: What is the central question of this study? An inverse correlation between circulating adiponectin and many diseases has been reported, but some studies have found no correlation. To evaluate this controversy, we investigated the relationship between heart-bound adiponectin and hypertension or cardiac hypertrophy, compared with serum adiponectin. What is the main finding and its importance? Using hypertensive and normotensive rats, we found that heart-bound adiponectin was inversely correlated with cardiac hypertrophy, suggesting that heart-bound adiponectin has a more important function in preventing cardiac hypertrophy than circulating adiponectin. Our study provides new insights regarding the role of adiponectin in diseases. The inverse correlation between circulating adiponectin concentration and hypertension or cardiac hypertrophy is still controversial. In addition to circulating adiponectin, adiponectin is also bound to tissues such as the heart and skeletal muscle. In this study, we investigated the relationship of serum adiponectin and heart-bound adiponectin with hypertension and cardiac hypertrophy. Four types of hypertensive rats presenting different blood pressure levels were used at different ages, as follows: normotensive Wistar-Kyoto rats (WKYs); two sub-strains (strains C and B2, having low and high blood pressure, respectively) of spontaneously hypertensive rats (SHRs); and stroke-prone SHRs (SHRSPs). Blood pressure, heart-to-body weight ratio, serum adiponectin and heart-bound adiponectin were determined. Histopathological analysis of the heart was carried out to evaluate the relationship with heart-bound adiponectin. Serum adiponectin concentration was not inversely correlated with blood pressure or heart-to-body weight ratio. In contrast, heart-bound adiponectin levels were significantly lower in SHRSPs than in other strains at respective ages. This resulted from a decrease in T-cadherin expression, which induced adiponectin binding to tissues. No significant difference in heart-bound adiponectin among WKYs and SHRs (C and B2) was detected, indicating that heart-bound adiponectin is not related to hypertension. In addition, differences in heart-bound adiponectin did not affect AMP-activated protein kinase in the traditional adiponectin activation cascade. Histopathological analysis revealed that heart-bound adiponectin was inversely correlated with cardiomyocyte hypertrophy and left ventricular wall thickness and, in part, with cardiac fibrosis. These results suggest that the decreased level of heart-bound adiponectin in SHRSPs is more related to their cardiac hypertrophy than circulating adiponectin.


Asunto(s)
Adiponectina/sangre , Hipertensión/sangre , Hipertrofia Ventricular Izquierda/sangre , Miocardio/metabolismo , Accidente Cerebrovascular/etiología , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adiponectina/genética , Factores de Edad , Animales , Biomarcadores/sangre , Presión Sanguínea , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Hipertensión/complicaciones , Hipertensión/fisiopatología , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Grasa Intraabdominal/metabolismo , Masculino , Miocardio/patología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Función Ventricular Izquierda , Remodelación Ventricular
4.
Clin Exp Pharmacol Physiol ; 44(2): 222-234, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27805296

RESUMEN

Statins induce apoptosis of tumour cells by inhibiting the prenylation of small G-proteins. However, the details of the apoptosis-inducing mechanisms remain poorly understood. The present study showed that the induction of apoptosis by statins in four different human head and neck squamous cell carcinoma (HNSCC) cell lines, HSC-3, HEp-2, Ca9-22, and SAS cells was mediated by increased caspase-3 activity. Statins induced apoptosis by the suppression of geranylgeranyl pyrophosphate biosynthesis. Furthermore, statins decreased the levels of phosphorylated ERK and mTOR by inhibiting the membrane localization of Ras and enhancing Bim expression in HSC-3 and HEp-2 cells. We also found that in all the cell types analyzed, the IC50 values for fluvastatin and simvastatin were highest in HEp-2 cells. In addition, HSC-3, Ca9-22, and SAS cells had higher Ras expression and membrane localization, higher activation of ERK1/2 and mTOR, and lower levels of Bim expression than HEp-2 cells. Our results indicate that statins induce apoptosis by increasing the activation of caspase-3 and by enhancing Bim expression through inhibition of the Ras/ERK and Ras/mTOR pathways. Furthermore, the sensitivity of HNSCC cells to statin treatment was closely related to Ras expression and prenylation levels, indicating that statins may act more effectively against tumours with high Ras expression and Ras-variability. Therefore, our findings support the use of statins as potential anticancer agents.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/metabolismo , Neoplasias de Cabeza y Cuello , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas ras/metabolismo , Proteína 11 Similar a Bcl2/genética , Caspasa 3/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Serina-Treonina Quinasas TOR/genética , Proteínas ras/genética
5.
Toxicol Appl Pharmacol ; 306: 105-12, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27417526

RESUMEN

Advanced metastatic melanoma, one of the most aggressive malignancies, is currently without reliable therapy. Therefore, new therapies are urgently needed. Mangiferin is a naturally occurring glucosylxanthone and exerts many beneficial biological activities. However, the effect of mangiferin on metastasis and tumor growth of metastatic melanoma remains unclear. In this study, we evaluated the effect of mangiferin on metastasis and tumor growth in a mouse metastatic melanoma model. We found that mangiferin inhibited spontaneous metastasis and tumor growth. Furthermore, mangiferin suppressed the nuclear translocation of nuclear factor kappa B (NF-κB) and expression of phosphorylated NF-κB-inducing kinase (NIK), inhibitor of kappa B kinase (IKK), and inhibitor of kappa B (IκB) and increases the expression of IκB protein in vivo. In addition, we found that mangiferin inhibited the expression of matrix metalloproteinases (MMPs) and very late antigens (VLAs) in vivo. Mangiferin treatment also increased the expression of cleaved caspase-3, cleaved Poly ADP ribose polymerase-1 (PARP-1), p53 upregulated modulator of apoptosis (PUMA), p53, and phosphorylated p53 proteins, and decreased the expression of Survivin and Bcl-associated X (Bcl-xL) proteins in vivo. These results indicate that mangiferin selectivity suppresses the NF-κB pathway via inhibition of NIK activation, thereby inhibiting metastasis and tumor growth. Importantly, the number of reported NIK selective inhibitors is limited. Taken together, our data suggest that mangiferin may be a potential therapeutic agent with a new mechanism of targeting NIK for the treatment of metastatic melanoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Xantonas/uso terapéutico , Animales , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Integrinas/genética , Masculino , Metaloproteinasas de la Matriz/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Xantonas/farmacología , Quinasa de Factor Nuclear kappa B
6.
Tumour Biol ; 37(7): 9099-110, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26762414

RESUMEN

Interaction between multiple myeloma (MM) cells and the bone marrow microenvironment plays a critical role in MM pathogenesis and the development of drug resistance. Recently, it has been reported that MM cells express the receptor activator of nuclear factor-κB (NF-κB) (RANK). However, the role of the RANK/RANK ligand (RANKL) system in drug resistance remains unclear. In this study, we demonstrated a novel function of the RANK/RANKL system in promoting drug resistance in MM. We found that RANKL treatment induced drug resistance in RANK-expressing but not RANK-negative cell lines. RANKL stimulation of RANK-expressing cells increased multidrug resistance protein 1 (MDR1), breast cancer resistance protein (BCRP), and lung resistance protein 1 (LRP1) expression and decreased Bim expression through various signaling molecules. RNA silencing of Bim expression induced drug resistance, but the RANKL-mediated drug resistance could not be overcome through the RNA silencing of MDR1, BCRP, and LRP1 expression. These results indicate that the RANK/RANKL system induces chemoresistance through the activation of multiple signal transduction pathways and by decreasing Bim expression in RANK-positive MM cells. These findings may prove to be useful in the development of cell adhesion-mediated drug resistance inhibitors in RANK-positive MM cells.


Asunto(s)
Adhesión Celular/genética , Resistencia a Medicamentos/genética , Mieloma Múltiple/genética , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteínas de Neoplasias/genética , Dominios y Motivos de Interacción de Proteínas/genética , Interferencia de ARN/fisiología , Transducción de Señal/genética
7.
Int J Cancer ; 137(1): 243-50, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25430564

RESUMEN

Oxaliplatin is a key drug commonly used in colorectal cancer treatment. Despite high clinical efficacy, its therapeutic application is limited by common, dose-limiting occurrence of neuropathy. As usual symptomatic neuropathy treatments fail to improve the patients' condition, there is an urgent need to advance our understanding of the pathogenesis of neuropathy to propose effective therapy and ensure adequate pain management. Oxaliplatin-induced neuropathy was recently reported to be associated with protein kinase C (PKC) activation. It is unclear, however, whether PKC inhibition can prevent neuropathy. In our current studies, we found that a PKC inhibitor, tamoxifen, inhibited oxaliplatin-induced neuropathy via the PKC/extracellular signal-regulated kinase (ERK)/c-Fos pathway in lumbar spinal cords (lumbar segments 4-6). Additionally, tamoxifen was shown to act in synergy with oxaliplatin to inhibit growth in tumor cells-implanted mice. Moreover, mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor, PD0325901, suppressed oxaliplatin-induced neuropathy and enhanced oxaliplatin efficacy. Our results indicate that oxaliplatin-induced neuropathy is associated with PKC/ERK/c-Fos pathway in lumbar spinal cord. Additionally, we demonstrate that disruption of this pathway by PKC and MEK inhibitors suppresses oxaliplatin-induced neuropathy, thereby suggesting that PKC and MEK inhibitors may be therapeutically useful in preventing oxaliplatin-induced neuropathy and could aid in combination antitumor pharmacotherapy.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Compuestos Organoplatinos/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Médula Espinal/efectos de los fármacos , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Difenilamina/administración & dosificación , Difenilamina/análogos & derivados , Difenilamina/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Hiperalgesia/inducido químicamente , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Compuestos Organoplatinos/efectos adversos , Oxaliplatino , Inhibidores de Proteínas Quinasas/farmacología , Médula Espinal/patología , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacología
8.
J Surg Oncol ; 111(8): 1041-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26060133

RESUMEN

BACKGROUND AND OBJECTIVES: We carried out a phase II trial to evaluate the feasibility, efficacy, and tolerability of perioperative chemotherapy including single intraperitoneal(IP) administration of paclitaxel(PTX) followed by intravenous(IV) administrations of PTX with S-1 in a neoadjuvant setting for serosa-positive gastric cancer. METHODS: Patients with cT4a gastric cancer were enrolled. A laparoscopic survey was performed before study inclusion for the confirmation of serosal invasion, negative lavage cytology, and negative peritoneal metastasis. IP PTX (80 mg/m(2)) was administered, followed by systemic chemotherapy. Surgery was performed after the completion of chemotherapy. The primary endpoint was the treatment completion rate. RESULTS: 37 patients were recruited. The treatment completion rate was 67.6% (25/37; 90% CI, 52.8-80.1%), which was significantly higher than 50%; we set this as a threshold value (P = 2.4% [one-sided]). 14 patients had target lesions; of these, 10 showed a partial response (71.4%), three had stable disease (21.4%), and one had progressive disease(7.2%). The response rate was 71.4% (10/14). All patients underwent gastrectomy with D2 lymph node dissection. The 3- and 5-year OS rates were 78.0 and 74.9%, respectively. CONCLUSIONS: Perioperative chemotherapy including neoadjuvant IP PTX followed by sequential IV PTX with S-1 for serosa-positive gastric cancer is feasible, safe, and efficient.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácido Oxónico/administración & dosificación , Paclitaxel/administración & dosificación , Neoplasias Gástricas/tratamiento farmacológico , Tegafur/administración & dosificación , Adulto , Anciano , Quimioterapia Adyuvante , Combinación de Medicamentos , Estudios de Factibilidad , Femenino , Gastrectomía , Humanos , Infusiones Intraventriculares , Infusiones Parenterales , Inyecciones Intraperitoneales , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante , Membrana Serosa/patología , Resultado del Tratamiento
9.
J Biomed Sci ; 21: 10, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24490900

RESUMEN

BACKGROUND: Bisphosphonates are an important class of antiresorptive drugs used in the treatment of metabolic bone diseases. Recent studies have shown that nitrogen-containing bisphosphonates induced apoptosis in rabbit osteoclasts and prevented prenylated small GTPase. However, whether bisphosphonates inhibit osteoclast formation has not been determined. In the present study, we investigated the inhibitory effect of minodronate and alendronate on the osteoclast formation and clarified the mechanism involved in a mouse macrophage-like cell lines C7 and RAW264.7. RESULTS: It was found that minodronate and alendronate inhibited the osteoclast formation of C7 cells induced by receptor activator of NF-κB ligand and macrophage colony stimulating factor, which are inhibited by the suppression of geranylgeranyl pyrophosphate (GGPP) biosynthesis. It was also found that minodronate and alendronate inhibited the osteoclast formation of RAW264.7 cells induced by receptor activator of NF-κB ligand. Furthermore, minodronate and alendornate decreased phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt; similarly, U0126, a mitogen protein kinase kinase 1/2 (MEK1/2) inhibitor, and LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, inhibited osteoclast formation. CONCLUSIONS: This indicates that minodronate and alendronate inhibit GGPP biosynthesis in the mevalonate pathway and then signal transduction in the MEK/ERK and PI3K/Akt pathways, thereby inhibiting osteoclast formation. These results suggest a novel effect of bisphosphonates that could be effective in the treatment of bone metabolic diseases, such as osteoporosis.


Asunto(s)
Alendronato/administración & dosificación , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Difosfonatos/administración & dosificación , Imidazoles/administración & dosificación , Osteoclastos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Difosfonatos/química , Humanos , Sistema de Señalización de MAP Quinasas/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Nitrógeno/química , Proteína Oncogénica v-akt/metabolismo , Fosfatos de Poliisoprenilo/biosíntesis , Ligando RANK/antagonistas & inhibidores
10.
J Nutr Sci Vitaminol (Tokyo) ; 70(1): 9-18, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38417857

RESUMEN

The development of atopic dermatitis (AD) involves multiple factors. Three such factors are particularly important in AD onset: immune abnormalities, skin barrier dysfunction, and itching. Many studies report that an imbalance between helper T (Th)1 and Th2 cells causes AD. Apple pectin, a prebiotic, has preventative effects in other allergic diseases (e.g., bronchial asthma and AD), but its potential benefits in AD are unclear. In this study, we investigated the effect of oral apple pectin administration on skin inflammation in an AD mouse model and examined changes in T cells involved in AD. To induce AD, a picryl chloride solution was applied to the shaved back skin of male NC/Nga mice. AD mice then received an oral apple pectin solution (0.4% or 4%) for 35 d. Compared with untreated AD mice, mice in both apple pectin-treated groups showed improvement in AD-induced inflammation and skin symptoms. Histological evaluation showed that apple pectin treatment attenuated epidermal thickening and decreased the number of mast cells and CD4+ cells in AD-induced mice. Apple pectin treatment also reduced serum IgE concentration, as well as expression of the inflammation indicator cyclooxygenase-2 and the Th2-related factors thymic stromal lymphopoietin, interleukin-4, and GATA3. Additionally, increased mRNA expression of the genes that encode interferon-γ and T-bet, which are Th1-related factors, and forkhead box protein P3, were observed in the apple pectin-treated groups. Our findings suggest that apple pectin treatment ameliorates AD by increasing regulatory T cells and improving the Th1/Th2 balance in the skin of AD model mice.


Asunto(s)
Dermatitis Atópica , Malus , Masculino , Ratones , Animales , Dermatitis Atópica/tratamiento farmacológico , Piel/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Administración Oral , Pectinas/farmacología , Pectinas/uso terapéutico , Modelos Animales de Enfermedad
11.
Cureus ; 16(6): e61522, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38957242

RESUMEN

Purpose The purpose of this study was to investigate the effect of dexmedetomidine (DEX) on hypotension-induced neuronal damage in a chronic cerebral hypoperfusion (CCH) model of rats, an established model of cerebral white matter lesions (WML) in humans, which is prevalent in the elderly and closely related to cognitive decline. Methods The CCH model rats were randomly assigned to one of four groups: normotension + no DEX (NN) group (n = 6), normotension + DEX (ND) group (n = 6), hypotension + no DEX (HN) group (n = 6), or hypotension + DEX (HD) group (n = 6). Under isoflurane anesthesia, mean arterial blood pressure was maintained at or above 80 mmHg (normotension) or below 60 mmHg (hypotension) for a duration of two hours. The DEX groups received 50 µg of DEX intraperitoneally. Two weeks later, the Y-maze test and, after preparing brain slices, immunohistochemical staining were performed using antibodies against neuronal nuclei (NeuN), microtubule-associated protein 2 (MAP2), glial fibrillary acidic protein (GFAP), and Ionized calcium-binding adapter molecule 1 (Iba1). Results Behavioral observations showed no significant differences among the groups. Significant reductions of both NeuN-positive cells and the MAP2-positive area were found in the hippocampal CA1 in the HN group compared with NN and ND groups, but not in the HD group. GFAP and Iba-1-positive areas were significantly increased in the HN group, but not in the HD group. Conclusion DEX significantly ameliorated hypotension-induced neuronal damage and both astroglial and microglial activation in the CA1 region of CCH rats.

12.
J Neural Transm (Vienna) ; 120(3): 361-74, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22955958

RESUMEN

We have previously reported free radical production after traumatic brain injury (TBI), which induces neural stem cell (NSC) degeneration and death. However, the effects of aging on NSC proliferation around the damaged area following TBI have not been investigated. Therefore, in this study, we used 10-week (young group) and 24-month-old (aged group) rat TBI models to investigate the effects of aging on NSC proliferation around damaged tissue using immunohistochemical and ex vivo techniques. Young and aged rats received TBI. At 1, 3 and 7 days after TBI, immunohistochemical and lipid peroxidation studies were performed. Immunohistochemistry revealed that the number of nestin-positive cells around the damaged area after TBI in the aged group decreased significantly when compared with those in the young group (P < 0.01). However, the number of 8-hydroxy-2'-deoxyguanosine-, 4-hydroxy-2-nonenal- and single-stranded DNA (ssDNA)-positive cells and the level of peroxidation around the damaged area after TBI significantly increased in the aged group, compared with those in the young group (P < 0.01). Furthermore, almost all ssDNA-positive cells in young and aged groups co-localized with NeuN and nestin staining. Ex vivo studies revealed that neurospheres, which differentiated into neurons and glia in culture, could only be isolated from injured brain tissue in young and aged groups at 3 days after TBI. These results indicate that, although there were fewer NSCs that have the potential to differentiate into neurons and glia, these NSCs escaped free radical-induced degeneration around the damaged area after TBI in the aged rat brain.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Peroxidación de Lípido/fisiología , Células-Madre Neurales/citología , 8-Hidroxi-2'-Desoxicoguanosina , Envejecimiento , Aldehídos/análisis , Animales , Diferenciación Celular/fisiología , ADN de Cadena Simple/análisis , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Desoxiguanosina/biosíntesis , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/análisis , Proteínas de Filamentos Intermediarios/biosíntesis , Masculino , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Células-Madre Neurales/metabolismo , Ratas , Ratas Wistar
13.
J Neural Transm (Vienna) ; 120(5): 767-83, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23180302

RESUMEN

Our previous study indicated that consuming (-)-epigallocatechin gallate (EGCG) before or after traumatic brain injury (TBI) eliminated free radical generation in rats, resulting in inhibition of neuronal degeneration and apoptotic death, and improvement of cognitive impairment. Here we investigated the effects of administering EGCG at various times pre- and post-TBI on cerebral function and morphology. Wistar rats were divided into five groups and were allowed access to (1) normal drinking water, (2) EGCG pre-TBI, (3) EGCG pre- and post-TBI, (4) EGCG post-TBI, and (5) sham-operated group with access to normal drinking water. TBI was induced with a pneumatic controlled injury device at 10 weeks of age. Immunohistochemistry and lipid peroxidation studies revealed that at 1, 3, and 7 days post-TBI, the number of 8-Hydroxy-2'-deoxyguanosine-, 4-Hydroxy-2-nonenal- and single-stranded DNA (ssDNA)-positive cells, and levels of malondialdehyde around the damaged area were significantly decreased in all EGCG treatment groups compared with the water group (P < 0.05). Although there was a significant increase in the number of surviving neurons after TBI in each EGCG treatment group compared with the water group (P < 0.05), significant improvement of cognitive impairment after TBI was only observed in the groups with continuous and post-TBI access to EGCG (P < 0.05). These results indicate that EGCG inhibits free radical-induced neuronal degeneration and apoptotic death around the area damaged by TBI. Importantly, continuous and post-TBI access to EGCG improved cerebral function following TBI. In summary, consumption of green tea may be an effective therapy for TBI patients.


Asunto(s)
Lesiones Encefálicas/prevención & control , Catequina/análogos & derivados , Fármacos Neuroprotectores/uso terapéutico , 8-Hidroxi-2'-Desoxicoguanosina , Aldehídos/metabolismo , Animales , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Catequina/uso terapéutico , ADN de Cadena Simple/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Proteína Ácida Fibrilar de la Glía/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
14.
Stroke ; 43(2): 470-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22156690

RESUMEN

BACKGROUND AND PURPOSE: There is an inverse association between dairy food consumption and the incidence of stroke in observational studies. However, it is unknown whether the relationship is causal or, if so, what components in milk are responsible for reducing the incidence of stroke. METHODS: Stroke-prone spontaneously hypertensive rats were fed diets comprising amino acids, proteins from different sources (casein, whey, soybean, or egg white), or fats from different sources (butter, beef tallow, or cocoa butter) and the onset of stroke and lifespan were examined. RESULTS: Increasing the amount of dietary casein (5% to 55% of caloric intake) markedly delayed the onset of stroke. However, when stroke-prone spontaneously hypertensive rats were fed diets containing 55% of caloric intake as protein, rats fed casein or whey protein, a major component of milk, displayed a delayed onset of stroke compared with rats fed soybean or egg white protein. Rats fed an amino acids diet containing the same amino acids composition as casein did not have a delay in the onset of stroke. Increasing dietary fats, including butter as well as beef tallow and cocoa butter, did not affect the onset of stroke. All diets did not affect blood pressure in the early stage. CONCLUSIONS: These data suggest that the inverse association between dairy food consumption and incidence of stroke in epidemiological studies is causal and that peptides in milk protein, but not fat, might be responsible for this effect.


Asunto(s)
Proteínas de la Leche/uso terapéutico , Accidente Cerebrovascular/prevención & control , Aminoácidos/farmacología , Animales , Presión Sanguínea/fisiología , Mantequilla , Caseínas/uso terapéutico , Hemorragia Cerebral/dietoterapia , Hemorragia Cerebral/patología , Hemorragia Cerebral/prevención & control , Infarto Cerebral/dietoterapia , Infarto Cerebral/patología , Infarto Cerebral/prevención & control , Dieta , Grasas de la Dieta/farmacología , Proteínas del Huevo/uso terapéutico , Masculino , Ratas , Ratas Endogámicas SHR , Proteínas de Soja/uso terapéutico , Glycine max/química , Accidente Cerebrovascular/dietoterapia , Accidente Cerebrovascular/patología , Urea/farmacología , Proteína de Suero de Leche
15.
Toxicol Appl Pharmacol ; 259(3): 402-10, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22326785

RESUMEN

Osteosarcoma is one of the most common primary malignant bone tumors in children and adolescents. Some patients continue to have a poor prognosis, because of the metastatic disease. YM529/ONO-5920 is a nitrogen-containing bisphosphonate that has been used for the treatment of osteoporosis. YM529/ONO-5920 has recently been reported to induce apoptosis in various tumors including osteosarcoma. However, the mode of metastasis suppression in osteosarcoma by YM529/ONO-5920 is unclear. In the present study, we investigated whether YM529/ONO-5920 inhibited tumor cell migration, invasion, adhesion, or metastasis in the LM8 mouse osteosarcoma cell line. We found that YM529/ONO-5920 significantly inhibited metastasis, cell migration, invasion, and adhesion at concentrations that did not have antiproliferative effects on LM8 cells. YM529/ONO-5920 also inhibited the mRNA expression and protein activities of matrix metalloproteinases (MMPs). In addition, YM529/ONO-5920 suppressed phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and the serine/threonine protein kinase B (Akt) by the inhibition of Ras prenylation. Moreover, U0126, a mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor, and LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, also inhibited LM8 cell migration, invasion, adhesion, and metastasis, as well as the mRNA expression and protein activities of MMP-1, MMP-2, MMP-9, and MT1-MMP. The results indicated that YM529/ONO-5920 suppressed the Ras/MEK/ERK and Ras/PI3K/Akt pathways, thereby inhibiting LM8 cell migration, invasion, adhesion, and metastasis. These findings suggest that YM529/ONO-5920 has potential clinical applications for the treatment of tumor cell metastasis in osteosarcoma.


Asunto(s)
Difosfonatos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Metástasis de la Neoplasia/prevención & control , Osteosarcoma/tratamiento farmacológico , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Masculino , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C3H , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Invasividad Neoplásica/prevención & control , Osteosarcoma/patología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Mensajero/metabolismo , Proteínas ras/antagonistas & inhibidores
16.
J Neural Transm (Vienna) ; 119(8): 877-90, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22212485

RESUMEN

A major component of green tea is (-)-epigallocatechin gallate (EGCG), which has strong antioxidant properties. Here, we investigated the effect of EGCG on neural stem cell (NSC) proliferation around the damaged area following traumatic brain injury (TBI). In this study, male Wistar rats that had access to normal drinking water, or water containing 0.1% (w/v) EGCG, ad libitum received TBI at 10 weeks of age. Immunohistochemistry revealed that the number of nestin-positive cells around the damaged area after TBI in the EGCG treatment group increased significantly compared with the normal water group (P < 0.05). However, the number of 8-hydroxy-2'-deoxyguanosine-, 4-hydroxy-2-nonenal-, single-stranded DNA (ssDNA)-positive cells and the level of peroxidation around the damaged area after TBI significantly decreased in the EGCG treatment group when compared with the water group (P < 0.05). Furthermore, in contrast to the EGCG group, almost all ssDNA-positive cells in the water group co-localized with NeuN and nestin-staining. Ex vivo studies revealed that spheres could only be isolated from injured brain tissue in the water group at 3 days following TBI. However, in the EGCG group, spheres could be isolated at both 3 and 7 days following TBI. A greater number of spheres could be isolated from the EGCG group, which differentiated into neurons and glia in culture without basic fibroblast growth factor. These results indicate that consumption of water containing EGCG pre- and post-TBI inhibits free radical-induced degradation of NSCs, which have the potential to differentiate into neurons and glia around the area of damage following TBI.


Asunto(s)
Lesiones Encefálicas/patología , Catequina/análogos & derivados , Peroxidación de Lípido/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Lesiones Encefálicas/metabolismo , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar
17.
J Surg Oncol ; 105(1): 43-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21837682

RESUMEN

BACKGROUND: The aim of this study was to examine the safety, pharmacokinetics, and cytological efficacy against free intraperitoneal cancer cells of intraperitoneal chemotherapy (IPC) with paclitaxel after gastrectomy with en-bloc D2 lymph node dissection (GD2) in cases of gastric cancer with peritoneal carcinomatosis (PC) and/or positive cytological findings in peritoneal washings (CFPW). METHODS: Twenty-one patients with gastric cancer with PC and/or positive CFPW who underwent GD2 were treated with early, post-operative, intraperitoneal paclitaxel. Intra-chemotherapeutic toxicity and operative complication were measured using the common toxicity criteria of the National Cancer Institute, version 3.0. Intraperitoneal and plasma paclitaxel concentrations were measured using a high-performance liquid chromatography assay. RESULTS: Grade 3 anemia occurred in two patients (9.5%) and neutropenia was observed in three patients (14.3%). No grade 4 toxicity was observed. A grade 2 operative complication was a superficial surgical site infection (4.8%) that was treated with antibiotics. Cytologically, no viable cancer cells were observed in the intra-abdominal fluid 24 hr after intraperitoneal administration of paclitaxel. The intraperitoneal/plasma area under the drug concentration-time curve (AUC) ratio was 596.9:1. CONCLUSION: IPC with paclitaxel after GD2 is a safe and cytologically effective treatment modality for free intraperitoneal cancer cells. However, additional data are required to determine the effect on survival.


Asunto(s)
Adenocarcinoma/terapia , Gastrectomía , Escisión del Ganglio Linfático , Paclitaxel/uso terapéutico , Neoplasias Peritoneales/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adulto , Anciano , Antineoplásicos Fitogénicos/uso terapéutico , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intraperitoneales , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/secundario , Complicaciones Posoperatorias , Estudios Prospectivos , Seguridad , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Tasa de Supervivencia , Resultado del Tratamiento
18.
Mediators Inflamm ; 2012: 701976, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23326018

RESUMEN

High blood levels of inflammatory biomarkers and immune cells in stroke lesions have been recognized as results of stroke. However, recent studies have suggested that inflammation occurs prior to stroke onset. In this study, we aimed to clarify the role of inflammation in stroke onset among stroke-prone spontaneously hypertensive rats (SHRSP). At 4 weeks of age (before stroke onset), the plasma level of IL-1ß was significantly higher in SHRSP (153.0 ± 49.7 pg/ml) than in Wistar Kyoto rats (WKY) (7.7 ± 3.4 pg/ml, P < 0.001 versus SHRSP) or spontaneously hypertensive rats (SHR) (28.0 ± 9.1 pg/ml, P < 0.001 versus SHRSP) (n = 6 per strain). Stimulated IL-1ß signal was also observed in cerebrovascular endothelial cells of SHRSP. Gene expressions of IL-1ß, IL-1 receptors, caspase-1, and downstream genes (MCP-1 and ICAM-1), which associated with immune cell recruitment, were significantly greater in SHRSP than in WKY or SHR, coincident with greater NFκB protein levels in SHRSP compared to WKY or SHR. In addition, continuous administration of IL-1ß (2 µg/day) using an osmotic pump slightly increased the incidence of stroke in SHR (P = 0.046) and significantly accelerated the onset of stroke in SHRSP (P = 0.006) compared to each control (n = 10 per group). These results suggest that a stimulated IL-1ß signal might be a cause of stroke onset when concomitant with severe hypertension.


Asunto(s)
Hipertensión/complicaciones , Inflamación/complicaciones , Interleucina-1beta/farmacología , Accidente Cerebrovascular/etiología , Animales , Presión Sanguínea , Peso Corporal , Masculino , Tamaño de los Órganos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
19.
Cytokine ; 54(1): 100-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21292498

RESUMEN

The tumor microenvironment plays a critical role in modulating malignant behavior and can dramatically influence cancer treatment strategies. We investigated whether statins inhibit the expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), and transforming growth factor-ß (TGF-ß) mRNA in the mouse osteosarcoma cell line LM8. We found that statins significantly inhibited mRNA expressions of bFGF, HGF, and TGF-ß, and bFGF, HGF, and TGF-ß secretions at concentrations that did not have antiproliferative effects on LM8 cells, but had no effect on the mRNA expression and secretion of VEGF. The inhibition of bFGF, HGF, and TGF-ß mRNA expression, and bFGF, HGF, TGF-ß secretions was reversed when geranylgeranyl pyrophosphate (GGPP), an intermediate in the mevalonate pathway, was used in combination with statins. Furthermore, statins reduced the membrane localization of K-Ras, phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated Akt. Our research indicates that statins inhibit GGPP biosynthesis in the mevalonate pathway, and then inhibit signal transduction in the Ras/ERK and Ras/Akt pathways, thereby inhibiting bFGF, HGF, TGF-ß expression in LM8 cells. These results suggest that statins are potentially useful as anti-angiogenic agents for the treatment of osteosarcoma.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica , Factor de Crecimiento de Hepatocito/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neovascularización Patológica , Osteosarcoma/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ras/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Ácido Mevalónico/metabolismo , Ratones , Fosforilación , Fosfatos de Poliisoprenilo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
J Neural Transm (Vienna) ; 118(2): 193-202, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20924619

RESUMEN

Exercise enhances neuronal stem cell (NSC) proliferation and neurogenesis. However, the effect of exercise on NSC proliferation surrounding the area of damage after traumatic brain injury (TBI) is unknown. Here, we investigate the effect of running on NSC proliferation following TBI in the rat. Wistar rats received TBI and were randomly divided into two groups: (1) non-exercise group and (2) exercise group. The exercise group ran on a treadmill for 30 min/day at 22 m/min for 7 consecutive days. Immunohistochemistry was used to monitor NSC proliferation around the damaged area, and ex vivo techniques were used to isolate NSCs from the damaged region in both groups. The number of nestin- and Ki67-positive cells observed at 3 and 7 days after TBI was significantly greater in the exercise group than in the non-exercise group (P < 0.01). Furthermore, most nestin-positive cells in the exercise group co-localized with Ki67-positive cells. In ex vivo studies, spheres could be isolated from injured brain tissue from the exercise group at 3 and 7 days following TBI, but at only 3 days in the non-exercise group. The number of spheres isolated from injured brain tissue was greater in the exercise group than in the non-exercise group. Spheres were immunopositive for nestin and comprised NSCs that could differentiate into neurons and glia. Exercise increases the proliferation of NSCs around the damaged area following TBI. Therefore, exercise therapy (rehabilitation) in the early phase following TBI is important for recuperation from cerebral dysfunction induced by TBI.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Condicionamiento Físico Animal/fisiología , Animales , Lesiones Encefálicas/patología , Diferenciación Celular/fisiología , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA