Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acid Ther ; 32(6): 486-496, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35895511

RESUMEN

Metabolic syndrome (MetS) is a pathological condition characterized by abdominal obesity, insulin resistance, hypertension, and hyperlipidemia. Sirtuin 1 (SIRT1), a highly conserved histone deacetylase, is characterized as a key metabolic regulator and protector against aging-associated pathologies, including MetS. In this study, we investigate the therapeutic potential of activating SIRT1 using small activating RNAs (saRNA), thereby reducing inflammatory-like responses and re-establishing normal lipid metabolism. SIRT1 saRNA significantly increased SIRT1 messenger RNA (mRNA) and protein levels in both lipopolysaccharide-stimulated and nonstimulated macrophages. SIRT1 saRNA significantly decreased inflammatory-like responses, by reducing mRNA levels of key inflammatory cytokines, such as Tumor Necrosis Factor alpha, Interleukin 1 beta (IL-1ß), Interleukin 6 (IL-6), and chemokines Monocyte Chemoattractant Protein-1 and keratinocyte chemoattractant. SIRT1 overexpression also significantly reduced phosphorylation of nuclear factor-κB and c-Jun N-terminal kinase, both key signaling molecules for the inflammatory pathway. To investigate the therapeutic effect of SIRT1 upregulation, we treated a high-fat diet model with SIRT1 saRNA conjugated to a transferrin receptor aptamer for delivery to the liver and cellular internalization. Animals in the SIRT1 saRNA treatment arm demonstrated significantly decreased weight gain with a significant reduction in white adipose tissue, triglycerides, fasting glucose levels, and intracellular lipid accumulation. These suggest treatment-induced changes to lipid and glucose metabolism in the animals. The results of this study demonstrate that targeted activation of SIRT1 by saRNAs is a potential strategy to reverse MetS.


Asunto(s)
Síndrome Metabólico , Humanos , Síndrome Metabólico/genética , Síndrome Metabólico/terapia , ARN Mensajero , Expresión Génica , Lípidos , Sirtuina 1/genética
2.
Adv Exp Med Biol ; 1313: 99-134, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34661893

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped, positive-sense RNA coronavirus responsible for the COVID-19 pandemic. Since December 2019, coronavirus disease 2019 (COVID-19) has affected more than 127 million people, 2.7 million deaths globally (as per WHO dashboard, dated 31 March, 2020), the virus is capable of transmitting from human to human via inhalation of infected respiratory droplets or aerosols or contact with infected fomites. Clinically, patients with COVID-19 present with severe respiratory distress syndrome, which is very similar to the presentation of other respiratory viral infections. A huge variation in the host response exists, with the resulting symptoms varying from mild to moderate. Comorbidities such as cardiovascular disease, hypertension, diabetes, coagulation dysfunction, stroke, malignant tumor and multiple organ dysfunction syndrome, as well as age and sex, are associated with severe COVID-19 cases. So far, no targeted therapies have been developed to treat this disease and existing drugs are being investigated for repurposing. This chapter discusses the epidemiology, clinical features of COVID-19, pathogenesis and the innate and adaptive immune response mounted by the host to the SARS-CoV-2 infection. A deeper understanding of the host-pathogen interaction is fundamental to the development of a vaccine.


Asunto(s)
COVID-19 , SARS-CoV-2 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Pandemias
4.
EBioMedicine ; 28: 31-42, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29398595

RESUMEN

Ischemia-reperfusion injury (IRI) during renal transplantation often initiates non-specific inflammatory responses that can result in the loss of kidney graft viability. However, the long-term consequence of IRI on renal grafts survival is uncertain. Here we review clinical evidence and laboratory studies, and elucidate the association between early IRI and later graft loss. Our critical analysis of previous publications indicates that early IRI does contribute to later graft loss through reduction of renal functional mass, graft vascular injury, and chronic hypoxia, as well as subsequent fibrosis. IRI is also known to induce kidney allograft dysfunction and acute rejection, reducing graft survival. Therefore, attempts have been made to substitute traditional preserving solutions with novel agents, yielding promising results.


Asunto(s)
Supervivencia de Injerto , Trasplante de Riñón , Daño por Reperfusión/terapia , Animales , Humanos , Modelos Biológicos
5.
Clin Cancer Res ; 23(13): 3453-3460, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986748

RESUMEN

Purpose: We aimed to establish whether programmed cell death-1 (PD-1) and programmed cell death ligand 1 (PD-L1) expression, in ovarian cancer tumor tissue and blood, could be used as biomarkers for discrimination of tumor histology and prognosis of ovarian cancer.Experimental Design: Immune cells were separated from blood, ascites, and tumor tissue obtained from women with suspected ovarian cancer and studied for the differential expression of possible immune biomarkers using flow cytometry. PD-L1 expression on tumor-associated inflammatory cells was assessed by immunohistochemistry and tissue microarray. Plasma soluble PD-L1 was measured using sandwich ELISA. The relationships among immune markers were explored using hierarchical cluster analyses.Results: Biomarkers from the discovery cohort that associated with PD-L1+ cells were found. PD-L1+ CD14+ cells and PD-L1+ CD11c+ cells in the monocyte gate showed a distinct expression pattern when comparing benign tumors and epithelial ovarian cancers (EOCs)-confirmed in the validation cohort. Receiver operating characteristic curves showed PD-L1+ and PD-L1+ CD14+ cells in the monocyte gate performed better than the well-established tumor marker CA-125 alone. Plasma soluble PD-L1 was elevated in patients with EOC compared with healthy women and patients with benign ovarian tumors. Low total PD-1+ expression on lymphocytes was associated with improved survival.Conclusions: Differential expression of immunological markers relating to the PD-1/PD-L1 pathway in blood can be used as potential diagnostic and prognostic markers in EOC. These data have implications for the development and trial of anti-PD-1/PD-L1 therapy in ovarian cancer. Clin Cancer Res; 23(13); 3453-60. ©2016 AACR.


Asunto(s)
Antígeno B7-H1/sangre , Biomarcadores de Tumor/sangre , Neoplasias Ováricas/sangre , Receptor de Muerte Celular Programada 1/sangre , Adulto , Anciano , Antígeno Ca-125/sangre , Antígeno Ca-125/inmunología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Pronóstico
6.
Adv Healthc Mater ; 4(8): 1180-9, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-25866054

RESUMEN

Adoptive T cell immunotherapy is a promising treatment strategy for epithelial ovarian cancer (EOC). However, programmed death ligand-1 (PD-L1), highly expressed on EOC cells, interacts with programmed death-1 (PD-1), expressed on T cells, causing immunosuppression. This study aims to block PD-1/PD-L1 interactions by delivering PD-L1 siRNA, using various folic acid (FA)-functionalized polyethylenimine (PEI) polymers, to SKOV-3-Luc EOC cells, and investigate the sensitization of the EOC cells to T cell killing. To enhance siRNA uptake into EOC cells, which over express folate receptors, PEI is modified with FA or PEG-FA so that siRNA is complexed into nanoparticles with folate molecules on the surface. PEI modification with a single functional group lowers the polymer cytotoxicity compared to unmodified PEI. FA-conjugated polymers increase siRNA uptake into SKOV-3-luc cells and decrease unspecific uptake into monocytes. All polymers result in 40% to 50% PD-L1 protein knockdown. Importantly, SKOV-3-Luc cells treated with either PEI-FA or PEI- polyethylene glycol (PEG)-FA/PD-L1 siRNA complexes are up to twofold more sensitive to T cell killing compared to scrambled siRNA treated controls. These findings are the first to demonstrate that PD-L1 knockdown in EOC cells, via siRNA/FA-targeted delivery, are able to sensitize cancer cells to T cell killing.


Asunto(s)
Antígeno B7-H1/genética , Ácido Fólico/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Polietileneimina/metabolismo , ARN Interferente Pequeño/genética , Antígeno B7-H1/metabolismo , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Terapia Genética , Humanos , Inmunoterapia/métodos , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Nanopartículas/química , Neoplasias Glandulares y Epiteliales/terapia , Neoplasias Ováricas/terapia , Tamaño de la Partícula , Polietilenglicoles/química , Polímeros/química , Linfocitos T/metabolismo
7.
Anesthesiology ; 122(6): 1312-26, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25856291

RESUMEN

BACKGROUND: Ischemia-reperfusion injury (IRI) of renal grafts may cause remote organ injury including lungs. The authors aimed to evaluate the protective effect of xenon exposure against remote lung injury due to renal graft IRI in a rat renal transplantation model. METHODS: For in vitro studies, human lung epithelial cell A549 was challenged with H2O2, tumor necrosis factor-α, or conditioned medium from human kidney proximal tubular cells (HK-2) after hypothermia-hypoxia insults. For in vivo studies, the Lewis renal graft was stored in 4°C Soltran preserving solution for 24 h and transplanted into the Lewis recipient, and the lungs were harvested 24 h after grafting. Cultured lung cells or the recipient after engraftment was exposed to 70% Xe or N2. Phospho (p)-mammalian target of rapamycin (mTOR), hypoxia-inducible factor-1α (HIF-1α), Bcl-2, high-mobility group protein-1 (HMGB-1), TLR-4, and nuclear factor κB (NF-κB) expression, lung inflammation, and cell injuries were assessed. RESULTS: Recipients receiving ischemic renal grafts developed pulmonary injury. Xenon treatment enhanced HIF-1α, which attenuated HMGB-1 translocation and NF-κB activation in A549 cells with oxidative and inflammatory stress. Xenon treatment enhanced p-mTOR, HIF-1α, and Bcl-2 expression and, in turn, promoted cell proliferation in the lung. Upon grafting, HMGB-1 translocation from lung epithelial nuclei was reduced; the TLR-4/NF-κB pathway was suppressed by xenon treatment; and subsequent tissue injury score (nitrogen vs. xenon: 26 ± 1.8 vs. 10.7 ± 2.6; n = 6) was significantly reduced. CONCLUSION: Xenon treatment confers protection against distant lung injury triggered by renal graft IRI, which is likely through the activation of mTOR-HIF-1α pathway and suppression of the HMGB-1 translocation from nuclei to cytoplasm.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Anestésicos por Inhalación/uso terapéutico , Trasplante de Riñón/efectos adversos , Xenón/uso terapéutico , Lesión Pulmonar Aguda/etiología , Animales , Línea Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/uso terapéutico , Inflamación/prevención & control , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/uso terapéutico , Ratas , Ratas Endogámicas Lew , Pruebas de Función Respiratoria , Transducción de Señal
8.
Am J Physiol Renal Physiol ; 306(8): F801-11, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24523386

RESUMEN

Toll-like receptor-4 (TLR-4) has been increasingly recognized as playing a critical role in the pathogenesis of ischemia-reperfusion injury (IRI) of renal grafts. This review provides a detailed overview of the new understanding of the involvement of TLR-4 in ischemia-reperfusion injury of renal grafts and its clinical significance in renal transplantation. TLR-4 not only responds to exogenous microbial motifs but can also recognize molecules which are released by stressed and necrotic cells, as well as degraded products of endogenous macromolecules. Upregulation of TLR-4 is found in tubular epithelial cells, vascular endothelial cells, and infiltrating leukocytes during renal ischemia-reperfusion injury, which is induced by massive release of endogenous damage-associated molecular pattern molecules such as high-mobility group box chromosomal protein 1. Activation of TLR-4 promotes the release of proinflammatory mediators, facilitates leukocyte migration and infiltration, activates the innate and adaptive immune system, and potentiates renal fibrosis. TLR-4 inhibition serves as the target of pharmacological agents, which could attenuate ischemia-reperfusion injury and associated delayed graft function and allograft rejection. There is evidence in the literature showing that targeting TLR-4 could improve long-term transplantation outcomes. Given the pivotal role of TLR-4 in ischemia-reperfusion injury and associated delayed graft function and allograft rejection, inhibition of TLR-4 using pharmacological agents could be beneficial for long-term graft survival.


Asunto(s)
Trasplante de Riñón/efectos adversos , Daño por Reperfusión/fisiopatología , Receptor Toll-Like 4/fisiología , Animales , Biglicano/fisiología , Rechazo de Injerto , Proteína HMGB1/fisiología , Proteínas de Choque Térmico/fisiología , Humanos , Inmunidad Innata/fisiología , Necrosis Tubular Aguda/etiología , Factor 88 de Diferenciación Mieloide/fisiología , Polimorfismo Genético , Transducción de Señal , Receptor Toll-Like 4/genética
9.
Kidney Int ; 85(1): 112-23, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24025645

RESUMEN

Chronic allograft nephropathy (CAN) is a common finding in kidney grafts with functional impairment. Prolonged hypothermic storage-induced ischemia-reperfusion injury is associated with the early onset of CAN. As the noble gas xenon is clinically used as an anesthetic and has renoprotective properties in a rodent model of ischemia-reperfusion injury, we studied whether early treatment with xenon could attenuate CAN associated with prolonged hypothermic storage. Exposure to xenon enhanced the expression of insulin growth factor-1 (IGF-1) and its receptor in human proximal tubular (HK-2) cells, which, in turn, increased cell proliferation. Xenon treatment before or after hypothermia-hypoxia decreased cell apoptosis and cell inflammation after reoxygenation. The xenon-induced HK-2 cell proliferation was abolished by blocking the IGF-1 receptor, mTOR, and HIF-1α individually. In the Fischer-to-Lewis rat allogeneic renal transplantation model, xenon exposure of donors before graft retrieval or recipients after engraftment enhanced tubular cell proliferation and decreased tubular cell death and cell inflammation associated with ischemia-reperfusion injury. Compared with control allografts, xenon treatment significantly suppressed T-cell infiltration and fibrosis, prevented the development of CAN, and improved renal function. Thus, xenon treatment promoted recovery from ischemia-reperfusion injury and reduced susceptibility to the subsequent development of CAN in allografts.


Asunto(s)
Anestésicos por Inhalación/uso terapéutico , Enfermedades Renales/prevención & control , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/prevención & control , Xenón/uso terapéutico , Animales , Línea Celular , Isquemia Fría/efectos adversos , Evaluación Preclínica de Medicamentos , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Masculino , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Receptor IGF Tipo 1/metabolismo , Daño por Reperfusión/etiología
10.
Cancer Immunol Immunother ; 63(3): 215-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24297569

RESUMEN

The programmed death-1 (PD-1) pathway is important in the maintenance of peripheral tolerance and homeostasis through suppression of T cell receptor signaling. As such, it is employed by many tumors as a means of immune escape. We have investigated the role of this pathway in human ovarian cancer (OC) to assess its potential role as a diagnostic and/or prognostic marker and therapeutic target, following recent clinical trial success of antibody therapy directed at this pathway. We show programmed death ligand-1 (PD-L1) expression on monocytes in the ascites and blood of patients with malignant OC is strikingly higher than those with benign/borderline disease, with no overlap in the values between these groups. We characterize the regulation of this molecule and show a role of IL-10 present in ascitic fluid. Flow cytometric analysis of T cells present in the ascites and blood showed a correlation of PD-1 expression with malignant tumors versus benign/borderline, in a similar manner to PD-L1 expression on monocytes. Finally, we demonstrate functional links between PD-L1 expression on monocytes and OC tumor cells with suppression of T cell responses. Overall, we present data based on samples obtained from women with ovarian cancer, suggesting the PD-1 pathway may be used as a reliable diagnostic marker in OC, as well as a viable target for use with PD-1/PD-L1-directed antibody immunotherapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/metabolismo , Terapia Molecular Dirigida/métodos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptor de Muerte Celular Programada 1/metabolismo , Anticuerpos Bloqueadores/inmunología , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Carcinogénesis , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia de Inmunosupresión , Interleucina-10/inmunología , Activación de Linfocitos , Monocitos/inmunología , Pronóstico , Linfocitos T/inmunología , Escape del Tumor
11.
FASEB J ; 27(12): 4822-33, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23934278

RESUMEN

Renal transplantation remains the best treatment option for patients with end-stage renal failure. However, the shortage of renal grafts remains a big challenge. Renal graft ischemic injuries that occur before and after graft retrieval have a devastating effect on graft survival, especially on grafts from marginal donors. This study was conducted to assess the protective effect against ischemic injury of a preservative solution supplemented with xenon (Xe), when used on ex vivo kidney grafts in a rat renal transplant model, and to explore the underlying mechanisms in vitro. Lewis rat renal grafts were stored in Soltran preservative solution at 4°C, saturated with nitrogen (N2) or Xe gas (70% Xe or N2, with 5% CO2 balanced with O2) for 24 or 48 h. Grafts stored in Xe-saturated preservative solution demonstrated significantly less severe histopathologic changes, together with enhanced B-cell lymphoma (Bcl)-2 and heat shock protein (HSP)-70 expression. After engraftment in the Lewis rat recipient, renal function was significantly improved in the Xe-treated grafts, and macrophage infiltration and fibrosis were reduced. Xe exposure enhanced Bcl-2 and HSP-70 expression in human renal tubular epithelial (HK-2) cells and prevented mitochondrial and nuclear damage. The release of the apoptogenic factors cytochrome c, apoptosis-inducing factor (AIF), and proinflammatory high-mobility group protein B1 (HMGB-1) was effectively suppressed. This study thus demonstrated for the first time that Xe confers renoprotection on renal grafts ex vivo and is likely to stabilize cellular structure during ischemic insult. The current study has significant clinical implications, in which the use of Xe ex vivo could enhance the marginal donor pool of renal grafts by preventing graft loss due to ischemia.


Asunto(s)
Trasplante de Riñón , Riñón/metabolismo , Preservación de Órganos/métodos , Xenón/farmacología , Animales , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/genética , Factor Inductor de la Apoptosis/metabolismo , Línea Celular , Citocromos c/genética , Citocromos c/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Riñón/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Endogámicas Lew
12.
Biomaterials ; 34(32): 7971-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23880339

RESUMEN

Hydrophobic modification of low molecular weight (LMW) polyethylenimine (PEI) is known to increase gene transfection efficiency of LMW PEI. However, few studies have explored how the conjugated hydrophobic groups influence the properties of the modified LMW PEI mainly due to difficulties in obtaining well defined final product compositions and limitations in current chemical synthesis routes. The aim of this study was to modify LMW PEI (Mn 1.8 kDa, PEI-1.8) judiciously with different hydrophobic functional groups and to investigate how hydrophobicity, molecular structure and inclusion of hydrogen bonding properties in the conjugated side groups as well as the conjugation degree (number of primary amine groups of PEI-1.8 modified with hydrophobic groups) influence PEI-1.8 gene transfection efficiency. The modified polymers were characterized for DNA binding ability, particle size, zeta potential, in vitro gene transfection efficiency and cytotoxicity in SKOV-3 human ovarian cancer and HepG2 human liver carcinoma cell lines. The study shows that modified PEI-1.8 polymers are able to condense plasmid DNA into cationic nanoparticles, of sizes ~100 nm, whereas unmodified polymer/DNA complexes display larger particle sizes of 2 µm. Hydrophobic modification also increases the zeta potential of polymer/DNA complexes. Importantly, modified PEI-1.8 shows enhanced transfection efficiency over the unmodified counterpart. Higher transfection efficiency is obtained when PEI-1.8 is modified with shorter hydrophobic groups (MTC-ethyl) as opposed to longer ones (MTC-octyl and MTC-deodecyl). An aromatic structured functional group (MTC-benzyl) also enhances transfection efficiency more than an alkyl functional group (MTC-octyl). An added hydrogen-bonding urea group in the conjugated functional group (MTC-urea) does not enhance transfection efficiency over one without urea (MTC-benzyl). The study also demonstrates that modification degree greatly influences gene transfection, and ~100% substitution of primary amine groups leads to significantly lower gene transfection efficiency. These findings provide insights to modification of PEI for development of effective and non-cytotoxic non-viral vectors.


Asunto(s)
Polietileneimina/química , Transfección/métodos , Línea Celular Tumoral , ADN/química , ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Vectores Genéticos/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Microscopía Confocal , Estructura Molecular , Peso Molecular , Nanopartículas/química , Tamaño de la Partícula , Plásmidos/química , Plásmidos/genética , Polímeros/química
13.
FASEB J ; 27(10): 4076-88, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23759444

RESUMEN

Prolonged hypothermic storage elicits severe ischemia-reperfusion injury (IRI) to renal grafts, contributing to delayed graft function (DGF) and episodes of acute immune rejection and shortened graft survival. Organoprotective strategies are therefore needed for improving long-term transplant outcome. The aim of this study is to investigate the renoprotective effect of xenon on early allograft injury associated with prolonged hypothermic storage. Xenon exposure enhanced the expression of heat-shock protein 70 (HSP-70) and heme oxygenase 1 (HO-1) and promoted cell survival after hypothermia-hypoxia insult in human proximal tubular (HK-2) cells, which was abolished by HSP-70 or HO-1 siRNA. In the brown Norway to Lewis rat renal transplantation, xenon administered to donor or recipient decreased the renal tubular cell death, inflammation, and MHC II expression, while delayed graft function (DGF) was therefore reduced. Pathological changes associated with acute rejection, including T-cell, macrophage, and fibroblast infiltration, were also decreased with xenon treatment. Donors or recipients treated with xenon in combination with cyclosporin A had prolonged renal allograft survival. Xenon protects allografts against delayed graft function, attenuates acute immune rejection, and enhances graft survival after prolonged hypothermic storage. Furthermore, xenon works additively with cyclosporin A to preserve post-transplant renal function.


Asunto(s)
Frío/efectos adversos , Trasplante de Riñón/efectos adversos , Riñón/patología , Daño por Reperfusión/etiología , Xenón/farmacología , Animales , Línea Celular , Ciclosporina/administración & dosificación , Ciclosporina/farmacología , Regulación de la Expresión Génica/fisiología , Genes MHC Clase II , Proteínas HSP70 de Choque Térmico , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Masculino , FN-kappa B , Ratas , Ratas Endogámicas Lew , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Manejo de Especímenes
14.
Immunology ; 138(2): 157-64, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23121382

RESUMEN

Previously we have shown that indoleamine 2,3-dioxygenase (IDO) and the tryptophan metabolite, 3-hydroxykynurenine (3HK) can prolong corneal allograft survival. IDO modulates the immune response by depletion of the essential amino acid tryptophan by breakdown to kynurenines, which themselves act directly on T lymphocytes. The tryptophan metabolite analogue N-(3,4-dimethoxycinnamonyl) anthranilic acid (DAA, 'Tranilast') shares the anthranilic acid core with 3HK. Systemic administration of DAA to mice receiving a fully MHC-mismatched allograft of cornea or skin resulted in significant delay in rejection (median survival of controls 12 days, 13 days for cornea and skin grafts, respectively, and of treated mice 24 days (P < 0.0001) and 17 days (P < 0.03), respectively). We provide evidence that DAA-induced suppression of the allogeneic response, in contrast to that induced by tryptophan metabolites, was a result of cell cycle arrest rather than T-cell death. Cell cycle arrest was mediated by up-regulation of the cell cycle-specific inhibitors p21 and p15, and associated with a significant reduction in interleukin-2 production, allowing us to characterize a novel mechanism for DAA-induced T-cell anergy. Currently licensed as an anti-allergy drug, the oral bioavailability and safe therapeutic profile of DAA make it a candidate for the prevention of rejection of transplanted cornea and other tissues.


Asunto(s)
Antialérgicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Trasplante de Córnea , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Piel , Linfocitos T/inmunología , ortoaminobenzoatos/farmacología , Animales , Puntos de Control del Ciclo Celular/inmunología , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Anergia Clonal/efectos de los fármacos , Anergia Clonal/inmunología , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Supervivencia de Injerto/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interleucina-2/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T/patología , Trasplante Homólogo , Triptófano/inmunología
15.
Eur J Immunol ; 43(3): 734-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23212959

RESUMEN

Dendritic cell (DC) modification is a potential strategy to induce clinical transplantation tolerance. We compared two DC modification strategies to inhibit allogeneic T-cell proliferation. In the first strategy, murine DCs were transduced with a lentiviral vector expressing CTLA4-KDEL, a fusion protein that prevents surface CD80/86 expression by retaining the co-stimulatory molecules within the ER. In the second approach, DCs were transduced to express the tryptophan-catabolising enzyme IDO. CTLA4-KDEL-expressing DCs induced anergy in alloreactive T cells and generated both CD4(+) CD25(+) and CD4(+) CD25(-) Treg cells (with direct and indirect donor allospecificity and capacity for linked suppression) both in vitro and in vivo. In contrast, T-cell unresponsiveness induced by IDO(+) DCs lacked donor specificity. In the absence of any immunosuppressive treatment, i.v. administration of CTLA4-KDEL-expressing DCs resulted in long-term survival of corneal allografts only when the DCs were capable of indirect presentation of alloantigen. This study demonstrates the therapeutic potential of CTLA4-KDEL-expressing DCs in tolerance induction.


Asunto(s)
Trasplante de Córnea , Células Dendríticas/inmunología , Rechazo de Injerto/inmunología , Inmunomodulación , Tolerancia al Trasplante/inmunología , Traslado Adoptivo , Animales , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Anergia Clonal/inmunología , Células Dendríticas/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/genética , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Inmunomodulación/genética , Lentivirus/genética , Activación de Linfocitos/inmunología , Ratones , Oligopéptidos/inmunología , Fenotipo , Señales de Clasificación de Proteína , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Transducción Genética , Trasplante Homólogo
16.
Cytotherapy ; 14(3): 274-84, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22136295

RESUMEN

BACKGROUND AIMS: Human embryonic stem cell (hESC)-derived mesenchymal stromal cells (MSC) (hESC-MSC) are an alternative source of MSC to bone marrow (BM)-derived MSC (BM-MSC), which are being investigated in clinical trials for their immunomodulatory potential. hESC-MSC have the advantage of being consistent because each batch can be generated from hESC under defined conditions. In contrast, BM-MSC have a limited proliferative capacity. METHODS: The ability to suppress the proliferation of anti-CD3/CD28-stimulated CD4 (+) T cells by hESC-MSC was compared with adult BM-MSC and neonatal foreskin fibroblast (Fb). RESULTS: hESC-MSC suppress the proliferation of CD4 (+) T cells in both contact and transwell systems, although inhibition is less in the transwell system. hESC-MSC are approximately 2-fold less potent (67 cells/100 T cells) than BM-MSC and Fb (37 and 34 cells/100 T cells, respectively) at suppressing T-cell proliferation by 50% in a transwell [inhibitory concentration(IC)(50)]. The anti-proliferative effect is not contact-dependent but requires the presence of factors such as interferon (IFN)-γ produced by activated T cells. IFN-γ induces the expression of indoleamine-2,3-dioxygenase (IDO) in hESC-MSC, BM-MSC and Fb, contributing to their immunosuppressive property. CONCLUSIONS: The feedback loop between MSC or Fb and activated T cells may limit the immunosuppressive effects of MSC and Fb to sites containing ongoing immunologic or inflammatory responses where activated T cells induce the up-regulation of IDO and immunomodulatory properties of MSC and Fb. These data demonstrate that hESC-MSC may be evaluated further as an allogeneic cell source for therapeutic applications requiring immunosuppression.


Asunto(s)
Células de la Médula Ósea/citología , Retroalimentación Fisiológica , Terapia de Inmunosupresión/métodos , Células Madre Mesenquimatosas/inmunología , Linfocitos T/inmunología , Adipocitos/citología , Adipocitos/inmunología , Células de la Médula Ósea/metabolismo , Antígenos CD28/inmunología , Complejo CD3/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/inmunología , Activación de Linfocitos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo
17.
Arthritis Rheum ; 63(12): 3758-67, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21953304

RESUMEN

OBJECTIVE: To isolate recombinant antibodies with specificity for human arthritic synovium and to develop targeting reagents with joint-specific delivery capacity for therapeutic and/or diagnostic applications. METHODS: In vivo single-chain Fv (scFv) antibody phage display screening using a human synovial xenograft model was used to isolate antibodies specific to the microvasculature of human arthritic synovium. Single-chain Fv antibody tissue-specific reactivity was assessed by immunostaining of synovial tissues from normal controls and from patients with rheumatoid arthritis and osteoarthritis, normal human tissue arrays, and tissues from other patients with inflammatory diseases displaying neovasculogenesis. In vivo scFv antibody tissue-specific targeting capacity was examined in the human synovial xenograft model using both (125)I-labeled and biotinylated antibody. RESULTS: We isolated a novel recombinant human antibody, scFv A7, with specificity for the microvasculature of human arthritic synovium. We showed that in vivo, this antibody could efficiently target human synovial microvasculature in SCID mice transplanted with human arthritic synovial xenografts. Our results demonstrated that scFv A7 antibody had no reactivity with the microvasculature or with other cellular components found in a comprehensive range of normal human tissues including normal human synovium. Further, we showed that the reactivity of the scFv A7 antibody was not a common feature of neovasculogenesis associated with chronic inflammatory conditions. CONCLUSION: Here we report for the first time the identification of an scFv antibody, A7, that specifically recognizes an epitope expressed in the microvasculature of human arthritic synovium and that has the potential to be developed as a joint-specific pharmaceutical.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Artritis Reumatoide/tratamiento farmacológico , Osteoartritis/tratamiento farmacológico , Anticuerpos de Cadena Única/uso terapéutico , Animales , Artritis Reumatoide/inmunología , Modelos Animales de Enfermedad , Epítopos/inmunología , Humanos , Ratones , Ratones SCID , Microvasos , Osteoartritis/inmunología , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Anticuerpos de Cadena Única/inmunología , Membrana Sinovial/irrigación sanguínea , Membrana Sinovial/inmunología , Trasplante Heterólogo
18.
J Nucl Med ; 52(9): 1441-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21852355

RESUMEN

UNLABELLED: The incidence and prevalence of gastroenteropancreatic neuroendocrine tumors has been increasing over the past 3 decades. Because of high densities of somatostatin receptors (sstr)--mainly sstr-2--on the cell surface of these tumors, (111)In-diethylenetriaminepentaacetic acid-octreotide scintigraphy has become an important part of clinical management. (18)F-radiolabeled analogs with suitable pharmacokinetics would permit PET with more rapid clinical protocols. METHODS: We compared the affinity in vitro and tissue pharmacokinetics by PET of 5 structurally related (19)F/(18)F-fluoroethyltriazole-Tyr(3)-octreotate (FET-TOCA) analogs: FET-G-polyethylene glycol (PEG)-TOCA, FETE-PEG-TOCA, FET-G-TOCA, FETE-TOCA, and FET-ßAG-TOCA to the recently described (18)F-aluminum fluoride NOTA-octreotide ((18)F-AIF-NOTA-OC) and the clinical radiotracer (68)Ga-DOTATATE. RESULTS: All (19)F-fluoroethyltriazole-Tyr(3)-octreotate compounds retained high agonist binding affinity to sstr-2 in vitro (half-maximal effective concentration, 4-19 nM vs. somatostatin at 5.6 nM). Dynamic PET showed that incorporation of PEG linkers, exemplified by (18)F-FET-G-PEG-TOCA and (18)F-FETE-PEG-TOCA, reduced uptake in high sstr-2-expressing AR42J pancreatic cancer xenografts. (18)F-FET-ßAG-TOCA showed the lowest nonspecific uptake in the liver. Tumor uptake increased in the order (68)Ga-DOTATATE < (18)F-AIF-NOTA ≤ (18)F-FET-ßAG-TOCA < (18)F-FET-G-TOCA. The uptake of (18)F-FET-ßAG-TOCA was specific: a radiolabeled scrambled peptide, (18)F-FET-ßAG-[W-c-(CTFTYC)K], did not show tumor uptake; there was lower uptake of (18)F-FET-ßAG-TOCA in AR42J xenografts when mice were pretreated with 10 mg of unlabeled octreotide per kilogram; and there was low uptake of (18)F-FET-ßAG-TOCA in low sstr-2-expressing HCT116 xenografts. CONCLUSION: We have developed novel fluoroethyltriazole-Tyr(3)-octreotate radioligands that combine high specific binding with rapid target localization and rapid pharmacokinetics for high-contrast PET. (18)F-FET-ßAG-TOCA and (18)F-FET-G-TOCA are candidates for future clinical evaluation.


Asunto(s)
Octreótido/análogos & derivados , Radiofármacos , Receptores de Somatostatina/metabolismo , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neoplasias Experimentales/diagnóstico por imagen , Octreótido/síntesis química , Octreótido/farmacocinética , Compuestos Organometálicos , Polietilenglicoles/química , Tomografía de Emisión de Positrones/métodos , Radiofármacos/síntesis química , Radiofármacos/farmacocinética , Relación Estructura-Actividad , Especificidad por Sustrato , Distribución Tisular
19.
Eur J Immunol ; 41(10): 2997-3005, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21805470

RESUMEN

The cornea is an immune privileged tissue. Since arginase has been found to modulate T-cell function by depleting arginine, we investigated the expression of arginase in the cornea and its possible role in immune privilege using a murine transplant model. We found that both the endothelium and epithelium of murine corneas express functional arginase I, capable of down-regulating T-cell proliferation in an in vitro culture system. The administration of the specific arginase inhibitor N-hydroxy-nor-L-Arg to recipient mice resulted in an accelerated rejection of allogeneic C57BL/6 (B6) corneal grafts. In contrast, in vivo blockade of arginase activity had no effect in altering the course of rejection of primary skin grafts that express little, if any, arginase. In addition, the inhibition of arginase did not alter systemic T-cell proliferation. These data show that arginase is functional in the cornea and contributes to the immune privilege of the eye, and that modulation of arginase contributes to graft survival.


Asunto(s)
Arginasa/antagonistas & inhibidores , Arginina/metabolismo , Córnea/inmunología , Trasplante de Córnea , Supervivencia de Injerto , Animales , Arginasa/metabolismo , Arginina/administración & dosificación , Arginina/análogos & derivados , Arginina/farmacología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Endotelio Corneal/inmunología , Endotelio Corneal/metabolismo , Epitelio Corneal/inmunología , Epitelio Corneal/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Trasplante de Piel , Trasplante Homólogo
20.
Bioorg Med Chem Lett ; 21(10): 3122-7, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21458258

RESUMEN

A novel class of alkyne linked [Tyr(3)]octreotate analogues have been labelled by a copper catalysed azide-alkyne cycloaddition reaction (CuAAC) to form a 1,4-substituted triazole using the reagent [(18)F]2-fluoroethyl azide. An unexpected variability in reactivity during the CuAAC reaction was observed for each alkyne analogue which has been investigated. Two lead alkyne linked [Tyr(3)]octreotate analogues, G-TOCA (3a) and ßAG-TOCA (5a) have been identified to be highly reactive in the click reaction showing complete conversion to the [(18)F]2-fluoroethyl triazole linked [Tyr(3)]octreotate analogues FET-G-TOCA (3b) and FET-ßAG-TOCA (5b) under mild conditions and with short synthesis times (5 min at 20 °C). As well as ease of synthesis, in vitro binding to the pancreatic tumour AR42J cells showed that both FET-G-TOCA and FET-ßAG-TOCA have high affinity for the somatostatin receptor with IC(50) of 4.0±1.4, and 1.6±0.2 nM, respectively.


Asunto(s)
Química Clic , Radioisótopos de Flúor , Péptidos Cíclicos/química , Radiofármacos/síntesis química , Triazoles/química , Catálisis , Cromatografía Líquida de Alta Presión , Cobre/química , Ciclización , Radioisótopos de Flúor/química , Estructura Molecular , Tomografía de Emisión de Positrones , Radiofármacos/química , Triazoles/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA