Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 89(5): 1062-1074, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27083283

RESUMEN

Activated Wnt signaling is critical in the pathogenesis of renal fibrosis, a final common pathway for most forms of chronic kidney disease. Therapeutic intervention by inhibition of individual Wnts or downstream Wnt/ß-catenin signaling has been proposed, but these approaches do not interrupt the functions of all Wnts nor block non-canonical Wnt signaling pathways. Alternatively, an orally bioavailable small molecule, Wnt-C59, blocks the catalytic activity of the Wnt-acyl transferase porcupine, and thereby prevents secretion of all Wnt isoforms. We found that inhibiting porcupine dramatically attenuates kidney fibrosis in the murine unilateral ureteral obstruction model. Wnt-C59 treatment similarly blunts collagen mRNA expression in the obstructed kidney. Consistent with its actions to broadly arrest Wnt signaling, porcupine inhibition reduces expression of Wnt target genes and bolsters nuclear exclusion of ß-catenin in the kidney following ureteral obstruction. Importantly, prevention of Wnt secretion by Wnt-C59 blunts expression of inflammatory cytokines in the obstructed kidney that otherwise provoke a positive feedback loop of Wnt expression in collagen-producing fibroblasts and epithelial cells. Thus, therapeutic targeting of porcupine abrogates kidney fibrosis not only by overcoming the redundancy of individual Wnt isoforms but also by preventing upstream cytokine-induced Wnt generation. These findings reveal a novel therapeutic maneuver to protect the kidney from fibrosis by interrupting a pathogenic crosstalk loop between locally generated inflammatory cytokines and the Wnt/ß-catenin signaling pathway.


Asunto(s)
Bencenoacetamidas/farmacología , Inhibidores Enzimáticos/farmacología , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Piridinas/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Acilación , Aciltransferasas , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/metabolismo , beta Catenina/metabolismo
2.
Gut Microbes ; 7(1): 48-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26939851

RESUMEN

Helicobacter pylori have been shown to influence physiological regulation of metabolic hormones involved in food intake, energy expenditure and body mass. It has been proposed that inducing H. pylori-induced gastric atrophy damages hormone-producing endocrine cells localized in gastric mucosal layers and therefore alter their concentrations. In a recent study, we provided additional proof in mice under controlled conditions that H. pylori and gut microbiota indeed affects circulating metabolic gut hormones and energy homeostasis. In this addendum, we presented data from follow-up investigations that demonstrated H. pylori and gut microbiota-associated modulation of metabolic gut hormones was independent and precedes H. pylori-induced histopathological changes in the gut of H. pylori-infected mice. Thus, H. pylori-associated argumentation of energy homeostasis is not caused by injury to endocrine cells in gastric mucosa.


Asunto(s)
Metabolismo Energético , Mucosa Gástrica/metabolismo , Microbioma Gastrointestinal , Helicobacter pylori/metabolismo , Animales , Mucosa Gástrica/microbiología , Ghrelina/metabolismo , Infecciones por Helicobacter/microbiología , Humanos , Insulina/metabolismo , Leptina/metabolismo , Ratones , Péptido YY/metabolismo
3.
Exp Anim ; 65(1): 53-62, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26558540

RESUMEN

We previously found that deletion of the multifunctional factor ANP32B (a.k.a. SSP29, APRIL, PAL31, PHAPI2) resulted in a severe but strain-specific defect resulting in perinatal lethality. The difficulty in generating an adult cohort of ANP32B-deficient animals limited our ability to examine adult phenotypes, particularly cancer-related phenotypes. We bred the Anp32b-null allele into the BALB/c and FVB/N genetic background. The BALB/c, but not the FVB/N, background provided sufficient frequency of adult Anp32b-null (Anp32b(-/-)) animals. From these, we found no apparent oncogenic role for this protein in mammary tumorigenesis contrary to what was predicted based on human data. We also found runtism, pathologies in various organ systems, and an unusual clinical chemistry signature in the adult Anp32b(-/-) mice. Intriguingly, genome-wide single-nucleotide polymorphism analysis suggested that our colony retained an unlinked C57BL/6J locus at high frequency. Breeding this locus to homozygosity demonstrated that it had a strong effect on Anp32b(-/-) viability indicating that this locus contains a modifier gene of Anp32b with respect to development. This suggests a functionally important genetic interaction with one of a limited number of candidate genes, foremost among them being the variant histone gene H2afv. Using congenic breeding strategies, we have generated a viable ANP32B-deficient animal in a mostly pure background. We have used this animal to reliably exclude mouse ANP32B as an important oncogene in mammary tumorigenesis. Our further phenotyping strengthens the evidence that ANP32B is a widespread regulator of gene expression. These studies may also impact the choice of subsequent groups with respect to congenic breeding versus de novo zygote targeting strategies for background analyses in mouse genetics.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ratones Endogámicos BALB C/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Alelos , Animales , Animales Congénicos , Cruzamiento , Proteínas de Ciclo Celular/deficiencia , Femenino , Genes Modificadores/genética , Estudios de Asociación Genética , Histonas/genética , Homocigoto , Masculino , Neoplasias Mamarias Animales/genética , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/deficiencia , Proteínas Nucleares/deficiencia , Fenotipo , Polimorfismo de Nucleótido Simple
4.
Eur J Pharm Sci ; 83: 143-54, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26709081

RESUMEN

PURPOSE: Overexpression of epidermal growth factor receptor (EGFR) is observed in oral squamous cell carcinoma (OSCC) and is associated with increased proliferation, metastasis and therapeutic resistance. We aim to develop a novel drug delivery system comprised of a photosensitizer Chlorin e6 (Ce6) that is encapsulated in a viral envelope and tagged with anti-EGFR antibody to target OSCC. METHODS: Ce6 was encapsulated in both virosomes (Ce6-Vir) and virosomes tagged with anti-EGFR antibody (Ce6-Vir-EGFR'). In vitro studies were conducted to assess the cellular uptake and bioavailability of the photosensitizer in OSCC cells. Ce6 alone or in constructs was then administered in a hamster cheek pouch model and fluorescence imaging and spectroscopy was performed. RESULTS: In vitro results showed that the uptake of Ce6-Vir-EGFR' was lower than that for Ce6-Vir and Ce6 possibly due to its large size. Nevertheless, in vivo results showed significant tumor specificity of Ce6-Vir-EGFR' compared to Ce6. The tumor to normal mucosa ratio showed that Ce6-Vir-EGFR' can successfully target OSCC lesions and therefore shows potential for use in fluorescence diagnosis of OSCC. CONCLUSIONS: Both the virosome-Ce6 constructs were internalized by OSCC cells and successfully used for fluorescence imaging. Tagging with anti-EGFR antibody further improved the targeting ability toward OSCC.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Receptores ErbB/inmunología , Mucosa Bucal/diagnóstico por imagen , Neoplasias de la Boca/diagnóstico por imagen , Fármacos Fotosensibilizantes/administración & dosificación , Porfirinas/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Carcinógenos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Mejilla , Clorofilidas , Cricetinae , Fluorescencia , Humanos , Masculino , Microscopía Fluorescente , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Imagen Óptica , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacocinética , Porfirinas/farmacología , Virosomas
5.
Exp Hematol ; 44(3): 189-93.e2, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26706195

RESUMEN

Despite the success of BCR-ABL1 tyrosine kinase inhibitors in patients with chronic myeloid leukemia (CML), resistance to tyrosine kinase inhibitors remains a therapeutic challenge. One strategy used to overcome resistance is combination of existing BCR-ABL1 tyrosine kinase inhibitors with agents that target alternative pathways. We report that inhibition of isoprenylcysteine carboxylmethyltransferase (Icmt), a key enzyme in the protein prenylation pathway, with the selective inhibitor cysmethynil enhances the effect of BCR-ABL1 tyrosine kinase inhibitors in killing CML cells. Cysmethynil augments tyrosine kinase inhibitor-induced apoptosis in both BCR-ABL1 wild type and BCR-ABL1 kinase domain mutant-expressing cell lines. Importantly, the enhanced apoptosis observed with the combination of cysmethynil and imatinib is significant only in primary CML CD34+ progenitor cells, not normal cord blood progenitor cells. The combination was also selective in inhibiting colony formation in CML CD34+ cells. The enhanced apoptosis appears to be due to combination of immediate and persistent inhibition of MAPK signaling. Consistent with in vitro studies, cysmethynil and imatinib, in combination, enhance the in vivo effects of either drug used alone. We found that simultaneous inhibition of BCR-ABL1 and Icmt may represent a potential therapeutic strategy for CML.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Mesilato de Imatinib/farmacología , Indoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Proteína Metiltransferasas/antagonistas & inhibidores , Animales , Femenino , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/metabolismo
6.
Oncotarget ; 6(32): 33769-80, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26378050

RESUMEN

The use of BCR-ABL1 tyrosine kinase inhibitors (TKI) has led to excellent clinical responses in patients with chronic phase chronic myeloid leukemia (CML). However these inhibitors have been less effective as single agents in the terminal blast phase (BP). We show that pyrvinium, a FDA-approved anthelminthic drug, selectively targets BP-CML CD34+ progenitor cells. Pyrvinium is effective in inducing apoptosis, inhibiting colony formation and self-renewal capacity of CD34+ cells from TKI-resistant BP-CML patients, while cord blood CD34+ are largely unaffected. The effects of pyrvinium are further enhanced upon combination with dasatinib, a second generation BCR-ABL1 TKI. In a CML xenograft model pyrvinium significantly inhibits tumor growth as a single agent, with complete inhibition in combination with dasatinib. While pyrvinium has been shown to inhibit the Wnt/ß-catenin signalling pathway via activation of casein kinase 1α , we find its activity in CML is not dependent on this pathway. Instead, we show that pyrvinium localizes to mitochondria and induces apoptosis by inhibiting mitochondrial respiration. Our study suggests that pyrvinium is a useful addition to the treatment armamentarium for BP-CML and that targeting mitochondrial respiration may be a potential therapeutic strategy in aggressive leukemia.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Mitocondrias/metabolismo , Compuestos de Pirvinio/administración & dosificación , Adenosina Trifosfato/química , Animales , Antígenos CD34/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Crisis Blástica/metabolismo , Quinasa de la Caseína I/metabolismo , Línea Celular Tumoral , Proliferación Celular , Dasatinib/administración & dosificación , Dasatinib/uso terapéutico , Humanos , Concentración 50 Inhibidora , Células K562 , Ratones , Ratones SCID , Trasplante de Neoplasias , Fosforilación , Compuestos de Pirvinio/uso terapéutico , Interferencia de ARN , beta Catenina/metabolismo
7.
Sci Rep ; 5: 8731, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25736205

RESUMEN

Helicobacter pylori, is an invariably commensal resident of the gut microbiome associated with gastric ulcer in adults. In addition, these patients also suffered from a low grade inflammation that activates the immune system and thus increased shunting of energy to host defense mechanisms. To assess whether a H. pylori infection could affect growth in early life, we determined the expression levels of selected metabolic gut hormones in germ free (GF) and specific pathogen-free (SPF) mice with and without the presence of H. pylori. Despite H. pylori-infected (SPFH) mice display alteration in host metabolism (elevated levels of leptin, insulin and peptide YY) compared to non-infected SPF mice, their growth curves remained the same. SPFH mice also displayed increased level of eotaxin-1. Interestingly, GF mice infected with H. pylori (GFH) also displayed increased levels of ghrelin and PYY. However, in contrast to SPFH mice, GFH showed reduced weight gain and malnutrition. These preliminary findings show that exposure to H. pylori alters host metabolism early in life; but the commensal microbiota in SPF mice can attenuate the growth retarding effect from H. pylori observed in GF mice. Further investigations of possible additional side effects of H. pylori are highly warranted.


Asunto(s)
Metabolismo Energético , Infecciones por Helicobacter/metabolismo , Intestino Grueso/metabolismo , Hormonas Peptídicas/metabolismo , Animales , Índice de Masa Corporal , Quimiocina CCL11/metabolismo , Ghrelina/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , Humanos , Insulina/metabolismo , Intestino Grueso/microbiología , Leptina/metabolismo , Ratones Endogámicos C57BL , Microbiota/fisiología , Modelos Biológicos , Péptido YY/metabolismo , Organismos Libres de Patógenos Específicos , Aumento de Peso/fisiología
8.
J Biomed Opt ; 19(11): 116006, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25388810

RESUMEN

Microcirculation lesion is a common symptom of chronic liver diseases in the form of vasculature deformation and circulation alteration. In acute to chronic liver diseases such as biliary atresia, microcirculation lesion can have an early onset. Detection of microcirculation lesion is meaningful for studying the progression of liver disease. We have combined wide-field fluorescence microscopy and a laser speckle contrast technique to characterize hepatic microcirculation in vivo without labeling in a bile-duct ligation rat fibrosis model of biliary atresia. Through quantitative image analysis of four microcirculation parameters, we observed significant microcirculation lesion in the early to middle stages of fibrosis. This bimodal imaging method is useful to assess hepatic microcirculation lesion for the study of liver diseases.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Hígado/irrigación sanguínea , Microcirculación/fisiología , Microscopía/métodos , Animales , Atresia Biliar , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Curva ROC , Ratas
9.
PLoS Pathog ; 10(1): e1003887, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24465207

RESUMEN

To be able to colonize its host, invading Salmonella enterica serovar Typhimurium must disrupt and severely affect host-microbiome homeostasis. Here we report that S. Typhimurium induces acute infectious colitis by inhibiting peroxisome proliferator-activated receptor gamma (PPARγ) expression in intestinal epithelial cells. Interestingly, this PPARγ down-regulation by S. Typhimurium is independent of TLR-4 signaling but triggers a marked elevation of host innate immune response genes, including that encoding the antimicrobial peptide lipocalin-2 (Lcn2). Accumulation of Lcn2 stabilizes the metalloproteinase MMP-9 via extracellular binding, which further aggravates the colitis. Remarkably, when exposed to S. Typhimurium, Lcn2-null mice exhibited a drastic reduction of the colitis and remained protected even at later stages of infection. Our data suggest a mechanism in which S. Typhimurium hijacks the control of host immune response genes such as those encoding PPARγ and Lcn2 to acquire residence in a host, which by evolution has established a symbiotic relation with its microbiome community to prevent pathogen invasion.


Asunto(s)
Proteínas de Fase Aguda/inmunología , Colitis/inmunología , Evasión Inmune , Lipocalinas/inmunología , Proteínas Oncogénicas/inmunología , PPAR gamma/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Enfermedad Aguda , Proteínas de Fase Aguda/genética , Animales , Línea Celular , Colitis/genética , Colitis/microbiología , Colitis/patología , Humanos , Lipocalina 2 , Lipocalinas/genética , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética , PPAR gamma/genética , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Salmonella typhimurium/patogenicidad , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
10.
Nanomedicine (Lond) ; 9(11): 1665-79, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24294981

RESUMEN

AIM: The aim of this work was to develop a liposomal formulation to facilitate delivery of a synergistic safingol/C2-ceramide combination in the treatment of acute myeloid leukemia (AML). MATERIALS & METHODS: Liposomes were prepared using the extrusion method and the bioactive lipids were encapsulated passively. Drug concentrations were determined by liquid chromatography tandem mass spectrometry. Antileukemic activity was evaluated using human leukemic cell lines, patient samples and U937 leukemic xenograft models. RESULTS: A stable liposome formulation was developed to coencapsulate safingol and C2-ceramide at 1:1 molar ratio with >90% encapsulation efficiency. The liposomal safingol/C2-ceramide was effective in AML cell lines, patient samples and murine xenograft models of AML, compared with liposomal safingol or liposomal C2-ceramide alone despite a dose reduction of 33%. CONCLUSION: Our study provided proof-of-concept evidence to deliver synergistic combination of bioactive lipid to achieve complete remission in AML.


Asunto(s)
Antineoplásicos/química , Sistemas de Liberación de Medicamentos , Leucemia Mieloide Aguda/tratamiento farmacológico , Liposomas/química , Animales , Proliferación Celular , Supervivencia Celular , Ceramidas/química , Colesterol/química , Cromatografía Liquida , Femenino , Humanos , Concentración 50 Inhibidora , Leucemia Mieloide Aguda/metabolismo , Lípidos/química , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Fosfatidilcolinas/química , Esfingosina/análogos & derivados , Esfingosina/química , Espectrometría de Masas en Tándem , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Sci Transl Med ; 5(197): 197ra101, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23926199

RESUMEN

Aristolochic acid (AA), a natural product of Aristolochia plants found in herbal remedies and health supplements, is a group 1 carcinogen that can cause nephrotoxicity and upper urinary tract urothelial cell carcinoma (UTUC). Whole-genome and exome analysis of nine AA-associated UTUCs revealed a strikingly high somatic mutation rate (150 mutations/Mb), exceeding smoking-associated lung cancer (8 mutations/Mb) and ultraviolet radiation-associated melanoma (111 mutations/Mb). The AA-UTUC mutational signature was characterized by A:T to T:A transversions at the sequence motif A[C|T]AGG, located primarily on nontranscribed strands. AA-induced mutations were also significantly enriched at splice sites, suggesting a role for splice-site mutations in UTUC pathogenesis. RNA sequencing of AA-UTUC confirmed a general up-regulation of nonsense-mediated decay machinery components and aberrant splicing events associated with splice-site mutations. We observed a high frequency of somatic mutations in chromatin modifiers, particularly KDM6A, in AA-UTUC, demonstrated the sufficiency of AA to induce renal dysplasia in mice, and reproduced the AA mutational signature in experimentally treated human renal tubular cells. Finally, exploring other malignancies that were not known to be associated with AA, we screened 93 hepatocellular carcinoma genomes/exomes and identified AA-like mutational signatures in 11. Our study highlights an unusual genome-wide AA mutational signature and the potential use of mutation signatures as "molecular fingerprints" for interrogating high-throughput cancer genome data to infer previous carcinogen exposures.


Asunto(s)
Ácidos Aristolóquicos/efectos adversos , Carcinógenos/análisis , Genoma Humano/genética , Mutación/efectos de los fármacos , Mutación/genética , Animales , Ácidos Aristolóquicos/análisis , Carcinógenos/toxicidad , Línea Celular Tumoral , Humanos , Enfermedades Renales/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Mutágenos/análisis , Mutágenos/toxicidad , Neoplasias/genética , Empalme del ARN/genética , Neoplasias Urológicas/genética , Urotelio/patología
12.
Hum Gene Ther ; 24(5): 508-19, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23527815

RESUMEN

Liver fibrosis generates fibrotic foci with abundant activated hepatic stellate cells and excessive collagen deposition juxtaposed with healthy regions. Targeted delivery of antifibrotic therapeutics to hepatic stellate cells (HSCs) might improve treatment outcomes and reduce adverse effects on healthy tissue. We delivered the hepatocyte growth factor (HGF) gene specifically to activated hepatic stellate cells in fibrotic liver using vitamin A-coupled liposomes by retrograde intrabiliary infusion to bypass capillarized hepatic sinusoids. The antifibrotic effects of DsRed2-HGF vector encapsulated within vitamin A-coupled liposomes were validated by decreases in fibrotic markers in vitro. Fibrotic cultures transfected with the targeted transgene showed a significant decrease in fibrotic markers such as transforming growth factor-ß1. In rats, dimethylnitrosamine-induced liver fibrosis is manifested by an increase in collagen deposition and severe defenestration of sinusoidal endothelial cells. The HSC-targeted transgene, administered via retrograde intrabiliary infusion in fibrotic rats, successfully reduced liver fibrosis markers alpha-smooth muscle actin and collagen, accompanied by an increase in the expression of DsRed2-HGF near the fibrotic foci. Thus, targeted delivery of HGF gene to hepatic stellate cells increased the transgene expression at the fibrotic foci and strongly enhanced its antifibrotic effects.


Asunto(s)
Técnicas de Transferencia de Gen , Células Estrelladas Hepáticas , Factor de Crecimiento de Hepatocito/uso terapéutico , Cirrosis Hepática/genética , Cirrosis Hepática/terapia , Animales , Conductos Biliares , Dimetilnitrosamina/toxicidad , Regulación de la Expresión Génica , Terapia Genética , Factor de Crecimiento de Hepatocito/genética , Humanos , Liposomas/uso terapéutico , Hígado/patología , Cirrosis Hepática/inducido químicamente , Ratas , Transgenes
13.
Laryngoscope ; 122(8): 1809-14, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22752972

RESUMEN

OBJECTIVES/HYPOTHESIS: Mucosal closure following laryngeal microsurgery can be challenging due to limited access, and incisions are often left to heal without formal closure. This study explores the hypothesis that a bioabsorbable microclip can be developed as an alternative method to close mucosal incisions in laryngeal microsurgery. STUDY DESIGN: Animal study. METHODS: In vitro studies were performed to determine if a suitable clip could be designed using bioabsorbable materials. A porcine model was used to study behavior of the clip in vivo. RESULTS: We initially studied poly-ϵ-caprolactone but encountered difficulty creating a small clip with the necessary material strength. Using magnesium we were able to produce a clip sufficiently small to close vocal fold incisions. Magnesium is biocompatible, bioabsorbable, and malleable, and has been used to manufacture vascular stents and sutures. The magnesium microclip could be deployed rapidly using modified 2-mm micro-laryngeal cup forceps, which enable the clip to close in a circular shape. In vitro and in vivo tests showed the clip held securely to the vocal fold mucosa. Macroscopic and histologic studies showed no significant injury to the contralateral vocal fold. There was no evidence of lower airway injury after implanting clips into the lower airway. CONCLUSIONS: The bioabsorbable clip could be used to close selected incisions in laryngeal microsurgery. Currently we are working to further reduce the size of the clip and modify its bioabsorption properties to enable precise control of degradation.


Asunto(s)
Implantes Absorbibles , Mucosa Laríngea/cirugía , Magnesio , Microcirugia/instrumentación , Poliésteres , Instrumentos Quirúrgicos , Animales , Diseño de Equipo , Reacción a Cuerpo Extraño/patología , Mucosa Laríngea/patología , Polipropilenos , Colgajos Quirúrgicos , Suturas , Porcinos , Pliegues Vocales/patología , Pliegues Vocales/cirugía , Cicatrización de Heridas/fisiología
14.
Int J Nanomedicine ; 7: 739-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359453

RESUMEN

Intraperitoneal (IP) therapy with platinum (Pt)-based drugs has shown promising results clinically; however, high locoregional concentration of the drug could lead to adverse side effects. In this study, IP administration was coupled with a folate receptor-targeted (FRT) liposomal system, in an attempt to achieve intracellular delivery of the Pt-based drug carboplatin in order to increase therapeutic efficacy and to minimize toxicity. In vitro and in vivo activity of FRT carboplatin liposomes was compared with the activity of free drug and nontargeted (NT) carboplatin liposomes using FR-overexpressing IGROV-1 ovarian cancer cells as the model. Significant reduction in cell viability was observed with FRT liposomes, which, compared with the free drug, provided an approximately twofold increase in carboplatin potency. The increase in drug potency was correlated with significantly higher cellular accumulation of Pt resulting from FRT liposomal delivery. Further evaluation was conducted in mice bearing intraperitoneally inoculated IGROV-1 ovarian tumor xenografts. A superior survival rate (five out of six animals) was achieved in animals treated with FRT carboplatin liposomes, injected intraperitoneally with a dose of 15 mg/kg and following a schedule of twice-weekly administration for 3 weeks. In contrast, no survivors were observed in the free drug or NT carboplatin liposome groups. The presence of cancer cells in lung and liver tissues was observed in the saline, free carboplatin, and NT carboplatin liposome groups. However, there was no sign of cancer cells or drug-related toxicity detected in tissues from the animals treated with FRT carboplatin liposomes. The results of this study have demonstrated for the first time that the approach of coupling IP administration with FRT liposomal delivery could provide significantly improved therapeutic efficacy of carboplatin in the treatment of metastatic ovarian cancer.


Asunto(s)
Carboplatino/administración & dosificación , Transportadores de Ácido Fólico/metabolismo , Liposomas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Animales , Carboplatino/química , Carboplatino/farmacocinética , Línea Celular Tumoral , Femenino , Histocitoquímica , Humanos , Estimación de Kaplan-Meier , Liposomas/química , Liposomas/farmacocinética , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Control Release ; 160(2): 290-8, 2012 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-22100388

RESUMEN

Prognosis of patients with acute myeloid leukemia (AML) remains poor despite the use of first-line induction chemotherapy. Therefore, it is imperative to find effective treatment for AML patients. Safingol is a bioactive sphingolipid which has demonstrated promising in vitro anti-leukemic properties; however, translation into clinical use is hampered by its low water solubility and dose-limiting hemolysis. The present study is the first to describe a rationally designed liposome formulation of safingol and demonstrate the anti-leukemic potential using a panel of human AML cell lines and patient samples as well as a human xenograft model in SCID mice. Encapsulation efficiency of safingol into liposomes was approximately 100%, and the release of drug followed square-root-of-time release model. The presence of a transmembrane pH gradient completely abolished the biological activity of liposomal safingol. A positive zeta potential, which influenced cellular accumulation of liposomal safingol, was crucial to the anti-leukemic activity. Liposomal safingol was effective against a wide range of AML subtypes with minimal hemolytic toxicity, and was able to extend the median survival time of the U937-inoculated mice to 31 days as compared to 23 days by free drug. The increase in therapeutic efficacy could be related to the increase in systemic drug exposure as a result of liposome encapsulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia Experimental/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Esfingosina/análogos & derivados , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Femenino , Hemólisis/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Solubilidad , Esfingosina/administración & dosificación , Esfingosina/efectos adversos , Esfingosina/sangre , Esfingosina/uso terapéutico , Análisis de Supervivencia , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Ther ; 19(9): 1727-36, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21587210

RESUMEN

Vaccines that aim to expand tumor-specific CD8(+) T cells have yielded disappointing results in cancer patients although they showed efficacy in transplantable tumor mouse models. Using a system that more faithfully mimics a progressing cancer and its immunoinhibitory microenvironment, we here show that in transgenic mice, which gradually develop adenocarcinomas due to expression of HPV-16 E7 within their thyroid, a highly immunogenic vaccine expressing E7 only induces low E7-specific CD8(+) T-cell responses, which fail to affect the size of the tumors. In contrast, the same type of vaccine expressing E7 fused to herpes simplex virus (HSV)-1 glycoprotein D (gD), an antagonist of the coinhibitory B- and T-lymphocyte attenuator (BTLA)/CD160-herpes virus entry mediator (HVEM) pathways, stimulates potent E7-specific CD8(+) T-cell responses, which can be augmented by repeated vaccination, resulting in initial regression of even large tumor masses in all mice with sustained regression in more than half of them. These results indicate that active immunization concomitantly with blockade of the immunoinhibitory HVEM-BTLA/CD160 pathways through HSV-1 gD may result in sustained tumor regression.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia Activa/métodos , Neoplasias/terapia , Proteínas E7 de Papillomavirus/inmunología , Animales , Animales Modificados Genéticamente , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/inmunología , Neoplasias/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Glándula Tiroides/inmunología , Vacunación/métodos
17.
Blood ; 117(15): 3974-82, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21325603

RESUMEN

Catalytic domain variants of activated factor VII (FVIIa) with enhanced hemostatic properties are highly attractive for the treatment of bleeding disorders via gene-based therapy. To explore this in a hemophilic mouse model, we characterized 2 variants of murine activated FVII (mFVIIa-VEAY and mFVIIa-DVQ) with modified catalytic domains, based on recombinant human FVIIa (rhFVIIa) variants. Using purified recombinant proteins, we showed that murine FVIIa (mFVIIa) and variants had comparable binding to human and murine tissue factor (TF) and exhibited similar extrinsic coagulant activity. In vitro in the absence of TF, the variants showed a 6- to 17-fold enhanced proteolytic and coagulant activity relative to mFVIIa, but increased inactivation by antithrombin. Gene delivery of mFVIIa-VEAY resulted in long-term, effective hemostasis at 5-fold lower expression levels relative to mFVIIa in hemophilia A mice or in hemophilia B mice with inhibitors to factor IX. However, expression of mFVIIa-VEAY at 14-fold higher than therapeutic levels resulted in a progressive mortality to 70% within 6 weeks after gene delivery. These results are the first demonstration of the hemostatic efficacy of continuous expression, in the presence or absence of inhibitors, of a high-activity gene-based FVIIa variant in an animal model of hemophilia.


Asunto(s)
Dominio Catalítico/genética , Factor VIIa/genética , Terapia Genética/métodos , Hemofilia A/terapia , Hemostasis/fisiología , Animales , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Factor VIIa/química , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/mortalidad , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Plásmidos/genética , Estructura Terciaria de Proteína
18.
Radiat Res ; 174(3): 331-40, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20726728

RESUMEN

Photodynamic therapy (PDT) with low light fluence rate has rarely been studied in protocols that use short drug-light intervals and thus deliver illumination while plasma concentrations of photosensitizer are high, creating a prominent vascular response. In this study, the effects of light fluence rate on PDT response were investigated using motexafin lutetium (10 mg/kg) in combination with 730 nm light and a 180-min drug-light interval. At 180 min, the plasma level of photosensitizer was 5.7 ng/microl compared to 3.1 ng/mg in RIF tumor, and PDT-mediated vascular effects were confirmed by a spasmodic decrease in blood flow during illumination. Light delivery at 25 mW/cm(2) significantly improved long-term tumor responses over that at 75 mW/cm(2). This effect could not be attributed to oxygen conservation at low fluence rate, because 25 mW/cm(2) PDT provided little benefit to tumor hemoglobin oxygen saturation. However, 25 mW/cm(2) PDT did prolong the duration of ischemic insult during illumination and was correspondingly associated with greater decreases in perfusion immediately after PDT, followed by smaller increases in total hemoglobin concentration in the hours after PDT. Increases in blood volume suggest blood pooling from suboptimal vascular damage; thus the smaller increases after 25 mW/cm(2) PDT provide evidence of more widespread vascular damage, which was accompanied by greater decreases in clonogenic survival. Further study of low fluence rate as a means to improve responses to PDT under conditions designed to predominantly damage vasculature is warranted.


Asunto(s)
Metaloporfirinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Vasos Sanguíneos/metabolismo , Humanos , Neoplasias/irrigación sanguínea , Oxígeno/metabolismo
19.
Ultrasound Med Biol ; 36(5): 853-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20381952

RESUMEN

The goal of this murine investigation was to evaluate the effect of an antivascular ultrasound treatment on the growth of an implanted melanoma and the consequent survival rate. After the intravenous injection of 0.2 mL ultrasound contrast agent (Definity), therapy (n = 15) was performed on 1-mL tumors for 3 min with low-intensity continuous ultrasound (3 MHz; 2.4 +/- 0.1 W cm(-2) [I(SATA)]); control mice (n = 17) received a sham treatment. Mice were euthanized once the tumor had reached 3 mL, and then survival percentage vs. time curves were plotted. The median survival time (time for tumor to reach 3 mL) for the treated group was 23 d and for the control group was 18 d; the difference was statistically significant (p

Asunto(s)
Melanoma/patología , Melanoma/terapia , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Terapia por Ultrasonido/métodos , Animales , Línea Celular Tumoral , Ratones , Ratones Endogámicos C3H , Análisis de Supervivencia , Tasa de Supervivencia , Resultado del Tratamiento
20.
Blood ; 115(3): 510-8, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965654

RESUMEN

Proper thymocyte development is required to establish T-cell central tolerance and to generate naive T cells, both of which are essential for T-cell homeostasis and a functional immune system. Here we demonstrate that the loss of transcription factor Foxp1 results in the abnormal development of T cells. Instead of generating naive T cells, Foxp1-deficient single-positive thymocytes acquire an activated phenotype prematurely in the thymus and lead to the generation of peripheral CD4(+) T and CD8(+) T cells that exhibit an activated phenotype and increased apoptosis and readily produce cytokines upon T-cell receptor engagement. These results identify Foxp1 as an essential transcriptional regulator for thymocyte development and the generation of quiescent naive T cells.


Asunto(s)
Diferenciación Celular/genética , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Linfocitos T/fisiología , Timo/fisiología , Animales , Apoptosis/genética , Apoptosis/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteínas Represoras/genética , Linfocitos T/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA