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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Nucleic Acids Res ; 37(18): 6276-89, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19692581

RESUMEN

Gene transfer to the corneal endothelium has potential in preventing corneal transplant rejection. In this study, we transfected mouse corneal endothelial cells (MCEC) with a class of novel arginine-rich oligopeptides. The peptides featured a tri-block design and mediated reporter gene expression in MCEC more efficiently than the commercial polyethylenimine standard. The functionality of each block was demonstrated to critically influence the performance of the peptide. Results from confocal imaging and flow cytometry then showed that energy-dependent endocytosis was the dominant form of uptake and multiple pathways were involved. Additionally, uptake was strongly dependent on interactions with cell-surface heparan sulphate. Fluorescence resonance energy transfer studies revealed that the peptide/DNA entered cells as an associated complex and some will have dissociated by 8.5 h. Large-scale accumulation of uncondensed DNA within the nucleus can also be observed by 26 h. Finally, as a proof of biological relevance, we transfected MCEC with plasmids encoding for the functional indoleamine 2,3-dioxygenase (IDO) enzyme. We then demonstrated that the expressed IDO could catalyse the degradation of l-tryptophan, which in turn suppressed the growth of CD4+ T-cells in a proliferation assay.


Asunto(s)
Endotelio Corneal/metabolismo , Oligopéptidos/química , Transfección , Animales , Arginina/química , Núcleo Celular/química , Células Cultivadas , ADN/análisis , Expresión Génica , Genes Reporteros , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones
16.
Macromol Rapid Commun ; 31(13): 1170-4, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21590871

RESUMEN

The delivery of immunomodulatory genes into the cornea prior to transplantation is one promising strategy to improve graft survival rates. We recently reported a class of novel triblock oligopeptides that could mediate efficient gene transfer into corneal endothelial cells. Now these peptides are characterised further and it is show that they lack distinct secondary structures. Peptide complexes are also demonstrated to be weakly haemolytic and transfection efficiency is shown to be sensitive to several experimental conditions. SEM and FRET confocal images are used to study the particle morphology and to show that they condense their DNA cargo well.

17.
Transplantation ; 85(11): 1640-7, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18551072

RESUMEN

BACKGROUND: The expression of chemokines is central to the recruitment of inflammatory cells for graft rejection, and modulation of chemokine action is of potential in preventing graft rejection. We have examined chemokine expression in a murine model of corneal allograft rejection, and also determined the effect of expressing a broad acting chemokine antagonist, viral macrophage inflammatory protein II (vMIP II), on graft survival. METHOD: The expression of chemokines in a murine model of corneal transplantation was determined by real time RT-PCR and, in the case of regulated on activation normal T-cell expressed and secreted, by ELISA. The plasmid encoding the virally derived chemokine antagonist, vMIP II, was introduced into the corneal endothelial cells using a non-viral vector consisting of liposomes and transferrin. The expression and activity of vMIP II was determined by ELISA and functional assays, and the effect on graft survival noted. RESULTS: After allotransplantation, there was up-regulation of all 11 chemokines examined. After gene delivery, there was expression of active vMIP II for more than 14 days and considerable prolongation of graft survival. This was associated with a decrease in leukocyte infiltration of the stroma of the cells. CONCLUSION: As expected there was considerable up-regulation of chemokines during allograft rejection. The expression of vMIP II showed considerable prolongation of graft survival. This is the first time we have observed prolongation of graft survival after a non-viral (as opposed to viral) means of gene delivery and indicates the potential of interfering with chemokine action to prevent corneal graft failure.


Asunto(s)
Quimiocinas/genética , Trasplante de Córnea/fisiología , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Supervivencia de Injerto/genética , ARN/genética , Receptores de Quimiocina/antagonistas & inhibidores , Animales , Western Blotting , Quimiocinas CC , Modelos Animales de Enfermedad , Estudios de Seguimiento , Terapia Genética/métodos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Plásmidos , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Trasplante Homólogo
18.
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
19.
BMC Bioinformatics ; 8: 196, 2007 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-17565685

RESUMEN

BACKGROUND: Quantifying cell division and death is central to many studies in the biological sciences. The fluorescent dye CFSE allows the tracking of cell division in vitro and in vivo and provides a rich source of information with which to test models of cell kinetics. Cell division and death have a stochastic component at the single-cell level, and the probabilities of these occurring in any given time interval may also undergo systematic variation at a population level. This gives rise to heterogeneity in proliferating cell populations. Branching processes provide a natural means of describing this behaviour. RESULTS: We present a likelihood-based method for estimating the parameters of branching process models of cell kinetics using CFSE-labeling experiments, and demonstrate its validity using synthetic and experimental datasets. Performing inference and model comparison with real CFSE data presents some statistical problems and we suggest methods of dealing with them. CONCLUSION: The approach we describe here can be used to recover the (potentially variable) division and death rates of any cell population for which division tracking information is available.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular , Células Cultivadas/citología , Células Cultivadas/fisiología , Fluoresceínas , Interpretación de Imagen Asistida por Computador/métodos , Microscopía Fluorescente/métodos , Succinimidas , Algoritmos , Simulación por Computador , Modelos Biológicos
20.
Arterioscler Thromb Vasc Biol ; 26(3): 462-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16357316

RESUMEN

OBJECTIVE: Endothelium is an important target for gene therapy. We have investigated the effect of viral and nonviral vectors on the phenotype and function of endothelial cells (ECs) and developed methods to block any activation caused by these vectors. METHODS AND RESULTS: Transduction of ECs with viral vectors, including adenovirus, lentiviruses, and Moloney murine leukemia virus, can induce a pro-inflammatory phenotype. This activation was reduced when nonviral vectors were used. We demonstrate that after transduction there is upregulation of dsRNA-triggered antiviral and PI3K/Akt signaling pathway. Blockade of the NFkappaB, PI3-K, or PKR signaling pathways all operated to inhibit partially virally induced activation, and inhibition of both PKR and PI3-K pathways totally blocked EC activation. Furthermore, inhibition of IFN-alpha/beta in addition to PI3-K was effective at preventing EC activation. CONCLUSIONS: Viral vectors, although efficient at transducing ECs, result in their activation. Blockade of the signaling pathways involved in viral activation may be used to prevent such activation.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Endotelio Vascular/metabolismo , Terapia Genética/efectos adversos , Vectores Genéticos/inmunología , Transducción de Señal/inmunología , Vasculitis/etiología , Adenoviridae/genética , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Adhesión Celular/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Terapia Genética/métodos , Humanos , Lentivirus/genética , Virus de la Leucemia Murina de Moloney/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/efectos adversos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Vena Safena/citología , Células TH1/citología , Células TH1/inmunología , Transducción Genética , Vasculitis/inmunología , Vasculitis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA