Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3243-3252, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37249614

RESUMEN

Neurotoxicity and nephrotoxicity are the major dose-limiting factors for the clinical use of colistin against multidrug-resistant (MDR) Gram-negative bacteria. This study aimed to investigate the neurotoxic and nephrotoxic effects of colistin formulated with in-house synthesized sodium deoxycholate sulfate (SDCS) in a mouse model. Male mice C57BL/6 were randomly divided into four groups: control (saline solution), colistin (15 mg/kg/day), colistin:SDCS 1:1, and colistin:SDCS 1:2. In the colistin:SDCS treatment groups, the dosage was 15 mg/kg/day colistin equivalent; all mice were treated for 7 successive days. The thermal tolerance, body weight gain and organ weights were measured. The levels of serum blood urea nitrogen (BUN), creatinine (Cr), superoxide dismutase (SOD), and catalase (CAT) were assessed. Histopathological damages were assessed on mice organ. The colistin:SDCS formulations significantly improved thermal pain response of the mice comparable to the control group. The administration did not impair kidney function as evidence from BUN and Cr results; however, the oxidative stress biomarkers decreased in the colistin and colistin-SDCS treated mice. Several abnormalities were observed in the kidney, liver, spleen, and sciatic nerve tissues following colistin treatment, which indicated evidence of toxicity. The colistin-SDCS formulations were associated with less acute toxicity and fewer nephrotoxic and neurotoxic changes compared with the colistin alone group which indicated that SDCS attenuated colistin nephrotoxicity and neurotoxicity. This study highlights the potential application of colistin formulated with SDCS for safer clinical use against MDR Gram-negative bacteria.


Asunto(s)
Colistina , Síndromes de Neurotoxicidad , Ratones , Masculino , Animales , Colistina/toxicidad , Ácido Desoxicólico/farmacología , Ratones Endogámicos C57BL , Riñón , Síndromes de Neurotoxicidad/patología , Sulfatos/farmacología , Antibacterianos/farmacología
2.
PLoS One ; 18(4): e0284303, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37040355

RESUMEN

Probiotics provide health benefits in various aspects and are believed to modulate the immune system by balancing gut microbiota homeostasis, termed the "microbiota-immune axis". Recent evidence supports that several Lactobacillus strains possess glucose-lowering and anti-inflammatory effects in an animal model of type 1 diabetes (T1D). Although probiotics of Lacticaseibacillus paracasei SD1 (SD1) and Lacticaseibacillus rhamnosus SD11 (SD11) exert human oral health benefits by reducing harmful bacterial populations, their clinical application regarding hypoglycemic-related traits as well as the underlying mechanisms are still lacking. In this report, we used multiple low doses of streptozotocin (STZ)-induced diabetic BALB/c mice to explore the effects of SD1 and SD11 supplementation on the regulation of markers related to T1D. Experimental mice were randomly assigned into five groups, non-STZ + V, STZ + V, STZ + SD1, STZ + SD11, and STZ + SDM (mixture of SD1 and SD11), and physiological data were measured every week. Blood and pancreas samples were collected at 4- and 8-weeks. Our results indicate that supplementation with SD1, SD11, or SDM for 8 weeks significantly improved body weights, glycemic levels, glucose tolerance, insulin levels, and lipid profiles. Probiotic administration also preserved islet integrity and increased ß-cell mass in STZ-injected mice, as well as prevented infiltration of macrophages, CD4+, and CD8+ T cells into the islets. Significantly, SD1 and SD11 suppressed the levels of IL1-ß, TNF-α and IFN-γ and increased IL-10, which is concomitant with the inhibition of cleaved caspase 3, caspase 9, caspase 8, proapoptotic Bax, NF-κBp65, pSTAT1, and iNOS. Additionally, the survival ability of ß-cells was mediated by upregulated anti-apoptotic Bcl2. We conclude that SD1 and SD11 attenuate STZ-induced diabetic mice by stabilizing glycemic levels and reducing inflammation, thereby protecting ß-cells. Among the probiotic treatment groups, SD11 revealed the best results in almost all parameters, indicating its potential use for alleviating hyperglycemia-associated symptoms.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Lacticaseibacillus paracasei , Lacticaseibacillus rhamnosus , Probióticos , Humanos , Ratones , Animales , Estreptozocina , Lacticaseibacillus , Diabetes Mellitus Experimental/tratamiento farmacológico , Muerte Celular , Inflamación , Probióticos/farmacología , Glucosa/uso terapéutico
3.
Exp Ther Med ; 20(2): 1385-1396, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32742373

RESUMEN

Hyperglycemia-induced oxidative stress and inflammation are hallmarks of liver damage in diabetes mellitus. Accumulating evidence has demonstrated that Pluchea indica leaf ethanol extract (PILE) possesses strong antioxidant and anti-inflammatory properties. However, studies of its effects on liver damage in streptozotocin (STZ)-induced diabetic animals remain insufficient. To the best of our knowledge, the present study was the first to illustrate that PILE mitigated liver injury in STZ animals. Mice were first pretreated with PILE at either 50 mg/kg (PILE 50) or 100 mg/kg (PILE 100) 2 weeks prior to the induction of hyperglycemia by multiple low doses of STZ. The mice were then fed with PILE 50 or PILE 100 for 4 or 8 weeks, following which liver weight, pathological changes, oxidative stress parameters, inflammation-related markers and caspase-mediated apoptosis were measured at each time point. Untreated STZ mice exhibited abnormal increases in liver weight and severe pathological changes. However, PILE 100 reduced the severity of the STZ-induced diabetic phenotype at both time points. A significant decrease in the levels of superoxide dismutase and catalase, in addition to an increase in malondialdehyde, were observed in the livers of untreated STZ mice, all of which were significantly reversed by treatment with PILE 100 for 8 weeks. Western blot analysis revealed reduced levels of liver inflammatory markers, including interleukin-6, tumor necrosis factor-α, NF-κB p65, transforming growth factor-ß1 and protein kinase C following PILE 100 treatment. Additionally, changes in the levels of apoptotic markers indicated that PILE 100 significantly attenuated caspase-9 and -3 expression, whilst preserving that of the Bcl-2 protein. In conclusion, the present study revealed that PILE alleviates hyperglycemia-induced liver injury by normalizing the various mediators of oxidative stress, inflammation and apoptosis.

4.
Oncogene ; 39(22): 4358-4374, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32313227

RESUMEN

Only a small number of genes are bona fide oncogenes and tumor suppressors such as Ras, Myc, ß-catenin, p53, and APC. However, targeting these cancer drivers frequently fail to demonstrate sustained cancer remission. Tumor heterogeneity and evolution contribute to cancer resistance and pose challenges for cancer therapy due to differential genomic rearrangement and expression driving distinct tumor responses to treatments. Here we report that intratumor heterogeneity of Wnt/ß-catenin modulator δ-catenin controls individual cell behavior to promote cancer. The differential intratumor subcellular localization of δ-catenin mirrors its compartmentalization in prostate cancer xenograft cultures as result of mutation-rendered δ-catenin truncations. Wild-type and δ-catenin mutants displayed distinct protein interactomes that highlight rewiring of signal networks. Localization specific δ-catenin mutants influenced p120ctn-dependent Rho GTPase phosphorylation and shifted cells towards differential bFGF-responsive growth and motility, a known signal to bypass androgen receptor dependence. Mutant δ-catenin promoted Myc-induced prostate tumorigenesis while increasing bFGF-p38 MAP kinase signaling, ß-catenin-HIF-1α expression, and the nuclear size. Therefore, intratumor δ-catenin heterogeneity originated from genetic remodeling promotes prostate cancer expansion towards androgen independent signaling, supporting a neomorphism model paradigm for targeting tumor progression.


Asunto(s)
Adenocarcinoma/patología , Cateninas/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/patología , Transporte Activo de Núcleo Celular , Adenocarcinoma/genética , Animales , Cateninas/genética , Línea Celular Tumoral , Núcleo Celular/ultraestructura , ADN de Neoplasias/genética , Transición Epitelial-Mesenquimal/genética , Exones/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Genes myc , Xenoinjertos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Metástasis Linfática/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Transgénicos , Mutación , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Análisis de Secuencia de ADN , Fracciones Subcelulares/química , beta Catenina/fisiología , Proteínas de Unión al GTP rho/fisiología , Catenina delta
5.
Drug Deliv Transl Res ; 10(1): 59-69, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31368043

RESUMEN

Aerosol inhalation of amphotericin B (AmB) can be a clinically compliant way to administer the drug directly to the pulmonary route for treatment as well as prophylaxis of invasive pulmonary aspergillosis (IPA). We report aerosol formulation of AmB using sodium deoxycholate sulfate (SDCS), a lipid carrier synthesized in-house using natural precursor deoxycholic acid. In vitro toxicity was determined by MTT assay. Biodistribution and histopathology in rats were evaluated in targeted organs including the lungs, kidneys, spleen, and liver. No toxicity was observed when lung and kidney cells treated with AmB-SDCS formulations up to 8 µg/mL and minimal toxicity at higher concentration 16 µg/mL, while the Fungizone®-like formulation induced toxicity to lung and kidney cells with viability decreasing from 86 to 41% and 100 to 49%, respectively, when compared with an equivalent concentration of AmB-SDCS. Renal and hepatic markers were raised for Fungizone®-like formulation-treated rats but not for AmB-SDCS formulations following 7 days of regular dosing by intratracheal instillation. AmB concentrations were highest in the lungs (5.4-8.3 µg/g) which were well above minimum inhibitory concentration (MIC) of all Aspergillus species. Plasma concentration was also above MIC (> 2 µg/mL) for all AmB-SDCS formulations in comparison with Fungizone®-like formulation. No evidence of abnormal histopathology was observed in the lungs, liver, spleen, and kidneys for all AmB-SDCS formulations but was observed for the group treated with Fungizone®-like formulation. It is concluded that AmB-SDCS formulations can be efficiently administered via intratracheal instillation with no evidence of toxicity and may find great value in the treatment as well as prophylaxis of IPA through inhalation route.


Asunto(s)
Anfotericina B/administración & dosificación , Antifúngicos/administración & dosificación , Aspergillus/efectos de los fármacos , Ácido Desoxicólico/administración & dosificación , Células Epiteliales/citología , Anfotericina B/farmacocinética , Anfotericina B/toxicidad , Animales , Antifúngicos/farmacocinética , Antifúngicos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ácido Desoxicólico/farmacocinética , Ácido Desoxicólico/toxicidad , Combinación de Medicamentos , Composición de Medicamentos , Células Epiteliales/efectos de los fármacos , Humanos , Instilación de Medicamentos , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Células RAW 264.7 , Ratas , Distribución Tisular , Pruebas de Toxicidad
6.
Exp Ther Med ; 18(2): 1164-1174, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31316610

RESUMEN

Abnormalities in insulin hormone levels leads to a hyperglycemic condition of diabetic mellitus. Hyperglycemia seriously induces organ and system destructions. The excessive accumulation of collagen fiber deposits occurs in inflammatory and reorganization processes of chronic liver diseases in type I insulin-dependent diabetes. Regarding the research objective, glabridin (GLB), an active compound of licorice, was used as a daily supplement (40 mg/kg) in order to decrease hepatocyte destruction and collagen deposition in liver tissue of diabetic animals induced by streptozotocin. A total of 40 were randomly allocated to five groups (each, n=10), control, control treated with GLB (GLB), diabetic rats (DM) injected with single dose of streptozotocin (60 mg/kg) to induce a diabetic condition, diabetic rats receiving GLB (DM+GLB; 40 mg/kg) and diabetic rats treated with glibenclamide (DM+GL; 4 mg/kg). Characteristic histopathological changes in liver cells and tissues of rats were determined by Masson's trichrome staining and transmission electron microscopy (TEM). Western blotting was used to detect the expression of the key markers, collagen type I and fibronectin proteins. The histological investigation of liver tissue of the DM group revealed that the collagen fiber deposition was increased in the periportal, pericentral and perisinusoidal spaces compared with controls. Hepatocytes appeared as small and fragmented cells in TEM examination. Collagenization of the perisinusoidal space was recently demonstrated to represent a new aspect of the microvascular abnormalities and liver fibrosis. Healthy hepatocytes with round nucleus were observed following supplementation of glabridin. In addition, collagen fiber deposition was reduced in the area adjacent to the perisinusoidal space. The expression of collagen type I and fibronectin decreased strongly following glabridin supplementation in DM+GLB rats compared with DM rats, indicating that the hepatic tissue reorganization regained its normal morphology. These findings suggest that it may be beneficial to examine the role of glabridin as a therapeutic agent in diabetes treatment in future research.

7.
PLoS One ; 14(2): e0212133, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30779805

RESUMEN

Loss of ß-cell mass and function is a fundamental feature of pathogenesis for type 1 and type 2 diabetes. Increasing evidence indicates that apoptosis is one of the main mechanisms of ß-cell death in both types. Ethanolic extracts of Pluchea indica leaf (PILE) have been reported to possess blood glucose lowering actions in vivo. Nevertheless, further study is required to determine the underlying mechanisms. In this report, we have investigated the preventive effects of PILE on multiple low doses of streptozotocin (MLDS)-induced ß-cell apoptosis. Mice were pre-treated with PILE at 50 mg/kg (PILE 50) or 100 mg/kg (PILE 100) for 2 weeks before streptozotocin (STZ) stimulation, and the treatment continued for 4 or 8 weeks. Results revealed that PILE 100 mice exhibited improved blood biochemistry, maintained a higher body weight, had decreased hyperglycemia, and restored islet architectures compared to non-treated STZ mice. Significantly, PILE 100 decreased levels of inflammatory response markers interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interlukin1-ß (IL-1ß), concomitant with the inhibition of caspase-3, caspase-8, capsepase-9, phosphorylation of signal transducer and activator of transcription 1 (pSTAT1), nuclear factor-κBp65 (NF-κBp65), and inducible nitric oxide synthase (iNOS). Additionally, survival and proliferative ability of ß-cells was mediated by up-regulated Bcl-2 and Ki67, respectively. These results provide strong evidence that pretreatment with PILE 100 effectively attenuated STZ-induced diabetes-related symptoms and these effects could be associated with the inhibition of cytokine-induced ß-cell apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Asteraceae/química , Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Extractos Vegetales/farmacología , Hojas de la Planta/química , Animales , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Relación Dosis-Respuesta a Droga , Etanol/química , Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/uso terapéutico , Linfocitos T/efectos de los fármacos
8.
Int J Pharm ; 530(1-2): 279-290, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28764982

RESUMEN

A molecularly imprinted polymer nanoparticle (MIP) was prepared by integrating a mixed functional monomer into a highly cross-linked polymer. The nanosized insulin as a template transferred into the binding cavities, anchored functional monomer(s) that the insulin structure formed within free space of the molecular size region by MIP nanoparticles. The oral administration with the insulin-loaded MIP resulted in higher fluorescence intensity of rhodamine-labeled insulin into the epithelial cells. We observed the correlation between the lipophilic domains of dye over the affected areas of sites with the interplay of the intestinal epithelial layer on the different intestinal sections. And, the detection with guinea pig anti-insulin antibody followed by goat anti-guinea pig antibody clearly elicited the efficient insulin function in the necessary biological milieu. The root mean square roughness of the MIP indicated difference of the surface density, significantly lower compared with the polymer attributed to the protein-mucin uptake that efficiently promoted the insulin penetration. Eventually electron microscopy data of the conjugated biotin-gold nanoparticles showed the transport of insulin across the intestinal epithelium via transcellular pathway, and the development of the pancreatic ß cell in the streptozocin-induced diabetic rats. Histopathological observation exhibited no obvious toxic effect after orally treated with MIP loaded insulin (100mg/kg) daily for 14days compared to control group. The use of an insulin-loaded MIP was proven to be an effective therapeutic protein delivery through transmucosal oral route.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Células Epiteliales/efectos de los fármacos , Insulina/administración & dosificación , Impresión Molecular , Nanopartículas/ultraestructura , Animales , Células Cultivadas , Cobayas , Intestinos/citología , Polímeros , Ratas
9.
Mol Cancer ; 9: 304, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21106062

RESUMEN

BACKGROUND: Neuronal synaptic junction protein δ-catenin (CTNND2) is often overexpressed in prostatic adenocarcinomas but the mechanisms of its activation are unknown. To address this question, we studied the hypothesis that Hes1, human homolog of Drosophila Hairy and enhancer of split (Hes) 1, is a transcriptional repressor of δ-catenin expression and plays an important role in molecular carcinogenesis. RESULTS: We identified that, using a δ-catenin promoter reporter assay, Hes1, but not its inactive mutant, significantly repressed the upregulation of δ-catenin-luciferase activities induced by E2F1. Hes1 binds directly to the E-boxes on δ-catenin promoter and can reduce the expression of δ-catenin in prostate cancer cells. In prostate cancer CWR22-Rv1 and PC3 cell lines, which showed distinct δ-catenin overexpression, E2F1 and Hes1 expression pattern was altered. The suppression of Hes1 expression, either by γ-secretase inhibitors or by siRNA against Hes1, increased δ-catenin expression. γ-Secretase inhibition delayed S/G2-phase transition during cell cycle progression and induced cell shape changes to extend cellular processes in prostate cancer cells. In neuroendocrine prostate cancer mouse model derived allograft NE-10 tumors, δ-catenin showed an increased expression while Hes1 expression was diminished. Furthermore, E2F1 transcription was very high in subgroup of NE-10 tumors in which Hes1 still displayed residual expression, while its expression was only moderately increased in NE-10 tumors where Hes1 expression was completely suppressed. CONCLUSION: These studies support coordinated regulation of δ-catenin expression by both the activating transcription factor E2F1 and repressive transcription factor Hes1 in prostate cancer progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cateninas/metabolismo , Factor de Transcripción E2F1/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Cateninas/genética , Línea Celular Tumoral , Dipéptidos/farmacología , Factor de Transcripción E2F1/genética , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Técnicas In Vitro , Masculino , Ratones , Microscopía Fluorescente , Neoplasias de la Próstata/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción HES-1 , Catenina delta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA