Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
BMJ Open Diabetes Res Care ; 12(2)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38485229

RESUMEN

INTRODUCTION: Static incubation (static glucose-stimulated insulin secretion, sGSIS) is a measure of islet secretory function. The Stimulation Index (SI; insulin produced in high glucose/insulin produced in low glucose) is currently used as a product release criterion of islet transplant potency. RESEARCH DESIGN AND METHODS: Our hypothesis was that the Delta, insulin secreted in high glucose minus insulin secreted in low glucose, would be more predictive. To evaluate this hypothesis, sGSIS was performed on 32 consecutive human islet preparations, immobilizing the islets in a slurry of Sepharose beads to minimize mechanical perturbation. Simultaneous full-mass subrenal capsular transplants were performed in chemically induced diabetic immunodeficient mice. Logistic regression analysis was used to determine optimal cut-points for diabetes reversal time and the Fisher Exact Test was used to assess the ability of the Delta and the SI to accurately classify transplant outcomes. Receiver operating characteristic curve analysis was performed on cut-point grouped data, assessing the predictive power and optimal cut-point for each sGSIS potency metric. Finally, standard Kaplan-Meier-type survival analysis was conducted. RESULTS: In the case of the sGSIS the Delta provided a superior islet potency metric relative to the SI.ConclusionsThe sGSIS Delta value is predicitive of time to diabetes reversal in the full mass human islet transplant bioassay.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Humanos , Ratones , Animales , Secreción de Insulina , Glucosa/farmacología , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/fisiología , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Bioensayo
2.
Diabetes ; 72(11): 1641-1651, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37625134

RESUMEN

Extracellular (e)ATP, a potent proinflammatory molecule, is released by dying/damaged cells at the site of inflammation and is degraded by the membrane ectonucleotidases CD39 and CD73. In this study, we sought to unveil the role of eATP degradation in autoimmune diabetes. We then assessed the effect of soluble CD39 (sCD39) administration in prevention and reversal studies in NOD mice as well as in mechanistic studies. Our data showed that eATP levels were increased in hyperglycemic NOD mice compared with prediabetic NOD mice. CD39 and CD73 were found expressed by both α- and ß-cells and by different subsets of T cells. Importantly, prediabetic NOD mice displayed increased frequencies of CD3+CD73+CD39+ cells within their pancreata, pancreatic lymph nodes, and spleens. The administration of sCD39 into prediabetic NOD mice reduced their eATP levels, abrogated the proliferation of CD4+- and CD8+-autoreactive T cells, and increased the frequency of regulatory T cells, while delaying the onset of T1D. Notably, concomitant administration of sCD39 and anti-CD3 showed a strong synergism in restoring normoglycemia in newly hyperglycemic NOD mice compared with monotherapy with anti-CD3 or with sCD39. The eATP/CD39 pathway plays an important role in the onset of T1D, and its targeting might represent a potential therapeutic strategy in T1D.

4.
Diabetologia ; 62(7): 1237-1250, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31087105

RESUMEN

AIMS/HYPOTHESIS: Autoimmune attack against the insulin-producing beta cells in the pancreatic islets results in type 1 diabetes. However, despite considerable research, details of the type 1 diabetes immunopathology in situ are not fully understood mainly because of difficult access to the pancreatic islets in vivo. METHODS: Here, we used direct non-invasive confocal imaging of islets transplanted in the anterior chamber of the eye (ACE) to investigate the anti-islet autoimmunity in NOD mice before, during and after diabetes onset. ACE-transplanted islets allowed longitudinal studies of the autoimmune attack against islets and revealed the infiltration kinetics and in situ motility dynamics of fluorescence-labelled autoreactive T cells during diabetes development. Ex vivo immunostaining was also used to compare immune cell infiltrations into islet grafts in the eye and kidney as well as in pancreatic islets of the same diabetic NOD mice. RESULTS: We found similar immune infiltration in native pancreatic and ACE-transplanted islets, which established the ACE-transplanted islets as reliable reporters of the autoimmune response. Longitudinal studies in ACE-transplanted islets identified in vivo hallmarks of islet inflammation that concurred with early immune infiltration of the islets and preceded their collapse and hyperglycaemia onset. A model incorporating data on ACE-transplanted islet degranulation and swelling allowed early prediction of the autoimmune attack in the pancreas and prompted treatments to intercept type 1 diabetes. CONCLUSIONS/INTERPRETATION: The current findings highlight the value of ACE-transplanted islets in studying early type 1 diabetes pathogenesis in vivo and underscore the need for timely intervention to halt disease progression.


Asunto(s)
Diabetes Mellitus Tipo 1/diagnóstico por imagen , Animales , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/cirugía , Supervivencia de Injerto/fisiología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/cirugía , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD
5.
Diabetes ; 67(10): 2038-2053, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30065030

RESUMEN

Extracellular ATP (eATP) activates T cells by engaging the P2X7R receptor. We identified two loss-of-function P2X7R mutations that are protective against type 1 diabetes (T1D) and thus hypothesized that eATP/P2X7R signaling may represent an early step in T1D onset. Specifically, we observed that in patients with newly diagnosed T1D, P2X7R is upregulated on CD8+ effector T cells in comparison with healthy control subjects. eATP is released at high levels by human/murine islets in vitro in high-glucose/inflammatory conditions, thus upregulating P2X7R on CD8+ T cells in vitro. P2X7R blockade with oxidized ATP reduces the CD8+ T cell-mediated autoimmune response in vitro and delays diabetes onset in NOD mice. Autoreactive CD8+ T-cell activation is highly dependent upon eATP/P2X7R-mediated priming, while a novel sP2X7R recombinant protein abrogates changes in metabolism and the autoimmune response associated with CD8+ T cells. eATP/P2X7R signaling facilitates the onset of autoimmune T1D by fueling autoreactive CD8+ cells and therefore represents a novel targeted therapeutic for the disorder.


Asunto(s)
Adenosina Trifosfato/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Autoinmunidad/genética , Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/genética , Femenino , Citometría de Flujo , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Mutación/genética , Receptores Purinérgicos P2X7/genética
6.
PLoS One ; 13(5): e0196570, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29723228

RESUMEN

BACKGROUND: Type 1 and Type 2 diabetes mellitus (T1DM and T2DM) are caused by beta(ß)-cell loss and functional impairment. Identification of mechanisms of ß-cell death and therapeutic interventions to enhance ß-cell survival are essential for prevention and treatment of diabetes. Oxidative stress is a common feature of both T1DM and T2DM; elevated biomarkers of oxidative stress are detected in blood, urine and tissues including pancreas of patients with DM. Islet transplantation is a promising treatment for diabetes. However, exposure to stress (chemical and mechanical) and ischemia-reperfusion during isolation and transplantation causes islet loss by generation of reactive oxygen species (ROS). Human intracellular antioxidant enzymes and related molecules are essential defenses against ROS. Antioxidant enzyme levels including superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX) have been shown to be low in islet cells. However, little is known about the expression and function of antioxidant enzymes within islet cell subsets. We evaluated the expression of the key antioxidant enzymes in ß- and alpha(α)-cell and accessed effects of oxidative stress, islet isolation and transplantation on ß/α-cell ratio and viability in human islets. METHODS: Human pancreata from T1DM, T2DM and non-diabetic deceased donors were obtained and analyzed by confocal microscopy. Isolated islets were (I) transplanted in the renal sub-capsular space of streptozotocin-induced diabetic nude mice (in vivo bioassay), or (II) exposed to oxidative (H2O2) and nitrosative (NO donor) stress for 24 hrs in vitro. The ratio, % viability and death of ß- and α-cells, and DNA damage (8OHdG) were measured. RESULTS AND CONCLUSIONS: Catalase and GPX expression was much lower in ß- than α-cells. The ß/α-cell ratio fells significantly following islet isolation and transplantation. Exposure to oxidative stress caused a significantly lower survival and viability, with higher DNA damage in ß- than α-cells. These findings identified the weakness of ß-cell antioxidant capacity as a main cause of vulnerability to oxidative stress. Potential strategies to enhance ß-cell antioxidant capacity might be effective in prevention/treatment of diabetes.


Asunto(s)
Antioxidantes/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Animales , Catalasa/metabolismo , Recuento de Células , Supervivencia Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/patología , Femenino , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Glutatión Peroxidasa/metabolismo , Humanos , Técnicas In Vitro , Células Secretoras de Insulina/patología , Islotes Pancreáticos/patología , Trasplante de Islotes Pancreáticos/patología , Ratones , Ratones Desnudos , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
7.
Cell Metab ; 27(3): 549-558.e4, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514065

RESUMEN

Every animal species has a signature blood glucose level or glycemic set point. These set points are different, and the normal glycemic levels (normoglycemia) of one species would be life threatening for other species. Mouse normoglycemia can be considered diabetic for humans. The biological determinants of the glycemic set point remain unclear. Here we show that the pancreatic islet imposes its glycemic set point on the organism, making it the bona fide glucostat in the body. Moreover, and in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point. These findings affect transplantation and regenerative approaches to treat diabetes because restoring normoglycemia may require more than replacing only the beta cells. Furthermore, therapeutic strategies using glucagon receptor antagonists as hypoglycemic agents need to be reassessed, as they may reset the overall glucostat in the organism.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Animales , Diabetes Mellitus Experimental , Humanos , Hipoglucemiantes/farmacología , Trasplante de Islotes Pancreáticos , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Comunicación Paracrina , Receptores de Glucagón/antagonistas & inhibidores
9.
Med Eng Phys ; 48: 3-13, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28416198

RESUMEN

Cellular transplantation may treat several human diseases by replacing damaged cells and/or providing a local source of trophic factors promoting regeneration. We utilized human renal epithelial cells (hRECs) isolated from cadaveric donors as a cell model. For efficacious implementation of hRECs for treatment of kidney diseases, we evaluated a novel encapsulation strategy for immunoisolation of hRECs and lentiviral transduction of the Green Fluorescent Protein (GFP) as model gene for genetic engineering of hRECs to secrete desired trophic factors. In specific, we determined whether encapsulation through conformal coating and/or GFP transduction of hRECs allowed preservation of cell viability and of their trophic factor secretion. To that end, we optimized cultures of hRECs and showed that aggregation in three-dimensional spheroids significantly preserved cell viability, proliferation, and trophic factor secretion. We also showed that both wild type and GFP-engineered hRECs could be efficiently encapsulated within conformal hydrogel coatings through our fluid dynamic platform and that this resulted in further improvement of cell viability and trophic factors secretion. Our findings may lay the groundwork for future therapeutics based on transplantation of genetically engineered human primary cells for treatment of diseases affecting kidneys and potentially other tissues.


Asunto(s)
Ingeniería Celular , Trasplante de Células , Células Epiteliales/citología , Riñón/citología , Supervivencia Celular , Estudios de Factibilidad , Humanos , Medicina Regenerativa , Esferoides Celulares/citología
10.
Acta Biomater ; 49: 272-283, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27915019

RESUMEN

Islet transplantation is a promising therapy for Type 1 diabetes mellitus; however, host inflammatory and immune responses lead to islet dysfunction and destruction, despite potent systemic immunosuppression. Grafting of poly(ethylene glycol) (PEG) to the periphery of cells or tissues can mitigate inflammation and immune recognition via generation of a steric barrier. Herein, we sought to evaluate the complementary impact of islet PEGylation with a short-course immunotherapy on the survival of fully-MHC mismatched islet allografts (DBA/2 islets into diabetic C57BL/6J recipients). Anti-Lymphocyte Function-associated Antigen 1 (LFA-1) antibody was selected as a complementary, transient, systemic immune monotherapy. Islets were PEGylated via an optimized protocol, with resulting islets exhibiting robust cell viability and function. Following transplantation, a significant subset of diabetic animals receiving PEGylated islets (60%) or anti-LFA-1 antibody (50%) exhibited long-term (>100d) normoglycemia. The combinatorial approach proved synergistic, with 78% of the grafts exhibiting euglycemia long-term. Additional studies examining graft cellular infiltrates at early time points characterized the local impact of the transplant protocol on graft survival. Results illustrate the capacity of a simple polymer grafting approach to impart significant immunoprotective effects via modulation of the local transplant environment, while short-term immunotherapy serves to complement this effect. STATEMENT OF SIGNIFICANCE: We believe this study is important and of interest to the biomaterials and transplant community for several reasons: 1) it provides an optimized protocol for the PEGylation of islets, with minimal impact on the coated islets, which can be easily translated for clinical applications; 2) this optimized protocol demonstrates the benefits of islet PEGylation in providing modest immunosuppression in a murine model; 3) this work demonstrates the combinatory impact of PEGylation with short-course immunotherapy (via LFA-1 blockage), illustrating the capacity of PEGylation to complement existing immunotherapy; and 4) it suggests macrophage phenotype shifting as the potential mechanism for this observed benefit.


Asunto(s)
Supervivencia de Injerto/inmunología , Inmunoterapia , Trasplante de Islotes Pancreáticos , Polietilenglicoles/química , Animales , Antígenos de Histocompatibilidad/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Animales , Supervivencia Tisular , Trasplante Homólogo
11.
Diabetes ; 65(5): 1350-61, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26916086

RESUMEN

Transplantation of pancreatic islets is a therapeutic option to preserve or restore ß-cell function. Our study was aimed at developing a clinically applicable protocol for extrahepatic transplantation of pancreatic islets. The potency of islets implanted onto the omentum, using an in situ-generated adherent, resorbable plasma-thrombin biologic scaffold, was evaluated in diabetic rat and nonhuman primate (NHP) models. Intraomental islet engraftment in the biologic scaffold was confirmed by achievement of improved metabolic function and preservation of islet cytoarchitecture, with reconstitution of rich intrainsular vascular networks in both species. Long-term nonfasting normoglycemia and adequate glucose clearance (tolerance tests) were achieved in both intrahepatic and intraomental sites in rats. Intraomental graft recipients displayed lower levels of serum biomarkers of islet distress (e.g., acute serum insulin) and inflammation (e.g., leptin and α2-macroglobulin). Importantly, low-purity (30:70% endocrine:exocrine) syngeneic rat islet preparations displayed function equivalent to that of pure (>95% endocrine) preparations after intraomental biologic scaffold implantation. Moreover, the biologic scaffold sustained allogeneic islet engraftment in immunosuppressed recipients. Collectively, our feasibility/efficacy data, along with the simplicity of the procedure and the safety of the biologic scaffold components, represented sufficient preclinical testing to proceed to a pilot phase I/II clinical trial.


Asunto(s)
Materiales Biocompatibles , Diabetes Mellitus Experimental/cirugía , Hiperglucemia/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Páncreas Artificial , Andamios del Tejido , Animales , Materiales Biocompatibles/efectos adversos , Materiales Biocompatibles/química , Biomarcadores/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Estudios de Factibilidad , Femenino , Terapia de Inmunosupresión/efectos adversos , Islotes Pancreáticos/citología , Islotes Pancreáticos/ultraestructura , Trasplante de Islotes Pancreáticos/efectos adversos , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/patología , Macaca fascicularis , Masculino , Microscopía Electrónica de Rastreo , Epiplón , Páncreas Artificial/efectos adversos , Plasma/química , Plasma/metabolismo , Ratas Endogámicas Lew , Ratas Endogámicas WF , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Propiedades de Superficie , Trombina/efectos adversos , Trombina/química , Trombina/metabolismo , Ingeniería de Tejidos , Andamios del Tejido/efectos adversos , Andamios del Tejido/química , Trasplante Heterólogo/efectos adversos , Trasplante Heterotópico/efectos adversos , Trasplante Isogénico/efectos adversos
12.
Diabetes Care ; 39(1): 149-57, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26628416

RESUMEN

OBJECTIVE: To determine the safety and effects on insulin secretion of umbilical cord (UC) mesenchymal stromal cells (MSCs) plus autologous bone marrow mononuclear cell (aBM-MNC) stem cell transplantation (SCT) without immunotherapy in established type 1 diabetes (T1D). RESEARCH DESIGN AND METHODS: Between January 2009 and December 2010, 42 patients with T1D were randomized (n = 21/group) to either SCT (1.1 × 10(6)/kg UC-MSC, 106.8 × 10(6)/kg aBM-MNC through supraselective pancreatic artery cannulation) or standard care (control). Patients were followed for 1 year at 3-month intervals. The primary end point was C-peptide area under the curve (AUC(C-Pep)) during an oral glucose tolerance test at 1 year. Additional end points were safety and tolerability of the procedure, metabolic control, and quality of life. RESULTS: The treatment was well tolerated. At 1 year, metabolic measures improved in treated patients: AUCC-Pep increased 105.7% (6.6 ± 6.1 to 13.6 ± 8.1 pmol/mL/180 min, P = 0.00012) in 20 of 21 responders, whereas it decreased 7.7% in control subjects (8.4 ± 6.8 to 7.7 ± 4.5 pmol/mL/180 min, P = 0.013 vs. SCT); insulin area under the curve increased 49.3% (1,477.8 ± 1,012.8 to 2,205.5 ± 1,194.0 mmol/mL/180 min, P = 0.01), whereas it decreased 5.7% in control subjects (1,517.7 ± 630.2 to 1,431.7 ± 441.6 mmol/mL/180 min, P = 0.027 vs. SCT). HbA1c decreased 12.6% (8.6 ± 0.81% [70.0 ± 7.1 mmol/mol] to 7.5 ± 1.0% [58.0 ± 8.6 mmol/mol], P < 0.01) in the treated group, whereas it increased 1.2% in the control group (8.7 ± 0.9% [72.0 ± 7.5 mmol/mol] to 8.8 ± 0.9% [73 ± 7.5 mmol/mol], P < 0.01 vs. SCT). Fasting glycemia decreased 24.4% (200.0 ± 51.1 to 151.2 ± 22.1 mg/dL, P < 0.002) and 4.3% in control subjects (192.4 ± 35.3 to 184.2 ± 34.3 mg/dL, P < 0.042). Daily insulin requirements decreased 29.2% in only the treated group (0.9 ± 0.2 to 0.6 ± 0.2 IU/day/kg, P = 0.001), with no change found in control subjects (0.9 ± 0.2 to 0.9 ± 0.2 IU/day/kg, P < 0.01 vs. SCT). CONCLUSIONS: Transplantation of UC-MSC and aBM-MNC was safe and associated with moderate improvement of metabolic measures in patients with established T1D.


Asunto(s)
Trasplante de Médula Ósea , Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre Mesenquimatosas , Adolescente , Adulto , Péptido C/sangre , Niño , Preescolar , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Insulina/uso terapéutico , Secreción de Insulina , Masculino , Proyectos Piloto , Calidad de Vida , Trasplante Autólogo , Cordón Umbilical/citología , Adulto Joven
13.
Tissue Eng Part A ; 21(15-16): 2250-61, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26027872

RESUMEN

Inflammation is a significant detriment to the engraftment of cells and tissues, particularly for islet transplantation, where a low tolerance for the inflammatory milieu results in significant graft loss. Local treatment with anti-inflammatories, such as glucocorticoids, provides the benefits of site-targeted delivery with minimization of the broad side effects associated with systemic delivery. Polydimethylsiloxane (PDMS) is a flexible platform that is capable of providing sustained delivery of hydrophobic drugs. Here, we evaluated the capacity of PDMS constructs loaded with the anti-inflammatory glucocorticoid dexamethasone (Dex) to locally mitigate inflammation in islet grafts. Dex-PDMS constructs, fabricated in rod or disk geometries, demonstrated prolonged and sustained release at therapeutically relevant levels. In vitro, Dex-PDMS constructs inhibited endotoxin-induced human monocyte and macrophage activation, but they did not impair islet viability or function. Dex-PDMS rods, co-transplanted with islet-seeded scaffolds in a murine model, demonstrated suppression of host inflammatory responses during early- and late-phase engraftment, without significantly altering islet graft potency. The facile nature of these glucocorticoid-doped PDMS constructs allows for the optimization of targeted dose delivery with wide applicability in cell and tissue transplantation.


Asunto(s)
Dexametasona , Dimetilpolisiloxanos , Trasplante de Islotes Pancreáticos , Animales , Línea Celular Tumoral , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Dexametasona/química , Dexametasona/farmacocinética , Dexametasona/farmacología , Dimetilpolisiloxanos/química , Dimetilpolisiloxanos/farmacología , Humanos , Inflamación/tratamiento farmacológico , Masculino , Ratones
14.
Pharmacol Res ; 98: 76-85, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25814189

RESUMEN

We are living exciting times in the field of beta cell replacement therapies for the treatment of diabetes. While steady progress has been recorded thus far in clinical islet transplantation, novel approaches are needed to make cell-based therapies more reproducible and leading to long-lasting success. The multiple facets of diabetes impose the need for a transdisciplinary approach to attain this goal, by targeting immunity, promoting engraftment and sustained functional potency. We discuss herein the emerging technologies applied to this rapidly evolving field.


Asunto(s)
Diabetes Mellitus/cirugía , Trasplante de Islotes Pancreáticos/métodos , Animales , Bioingeniería , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 1/cirugía , Humanos , Inmunomodulación
15.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25786390

RESUMEN

With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.


Asunto(s)
Fibrina/química , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Humanos , Hidrogeles/química , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-sis/química , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/farmacología , Factor A de Crecimiento Endotelial Vascular/química
16.
Curr Med Chem ; 22(7): 891-901, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25312216

RESUMEN

Diabetes is a metabolic disorder characterized by elevation of glucose levels in the blood that develops in humans as a result of genetic predisposition and environmental factors. Unbalanced glycemic control has been associated with the development of progressive and debilitating complications that dramatically affect the quality of life and life expectancy of people with diabetes. The purinergic system represents a widely diffused signaling pathway in mammalian cells of different tissues where it plays critical roles in both physiological and pathological conditions. Herein we review the increasing evidence supporting that the purinergic system plays an important role in the multiple facets of diabetes, including its physiopathology and complications. We also discuss the potential relevance of the purinergic pathway for diagnosis and treatment of diabetes.


Asunto(s)
Diabetes Mellitus/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animales , Humanos , Transducción de Señal
17.
Pharmacogenomics ; 15(16): 2063-82, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25521362

RESUMEN

The most important goal in the treatment of patients with diabetes is to prevent the risk of cardiovascular disease (CVD), the first cause of mortality in these subjects. Thiazolidinediones (TZDs), a class of antidiabetic drugs, act as insulin sensitizers increasing insulin-dependent glucose disposal and reducing hepatic glucose output. TZDs including pioglitazone, rosiglitazone and troglitazone, by activating PPAR-γ have shown pleiotropic effects in reducing vascular risk factors and atherosclerosis. However, troglitazone was removed from the market due to its hepatoxicity, and rosiglitazone and pioglitazone both have particular warnings due to being associated with heart diseases. Specific genetic variations in genes involved in the pathways regulated by TDZs have demonstrated to modify the variability in treatment with these drugs, especially in their side effects. Therefore, pharmacogenomics and pharmacogenetics are an important tool in further understand intersubject variability per se but also to assess the therapeutic potential of such variability in drug individualization and therapeutic optimization.


Asunto(s)
Enfermedades Cardiovasculares/genética , Diabetes Mellitus/genética , Farmacogenética , Tiazolidinedionas/efectos adversos , Enfermedades Cardiovasculares/tratamiento farmacológico , Cromanos/efectos adversos , Cromanos/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Humanos , Pioglitazona , Medicina de Precisión , Factores de Riesgo , Rosiglitazona , Tiazolidinedionas/uso terapéutico , Troglitazona
18.
Methods Mol Biol ; 1186: 121-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25149307

RESUMEN

Intravital imaging approaches are proving to be essential to address new questions to better understand how the immune system operates. These approaches are especially valuable to characterize the complex organization of immune responses in vivo. Here, we examine how to take advantage of the cornea as a natural body window to apply noninvasive imaging techniques to assess cytotoxic T lymphocyte involvement in the immune rejection process in a model of intraocular allogeneic islet transplantation.


Asunto(s)
Cámara Anterior/inmunología , Rechazo de Injerto/inmunología , Trasplante de Islotes Pancreáticos/métodos , Microscopía/métodos , Linfocitos T Citotóxicos/inmunología , Animales , Córnea/fisiología , Citotoxicidad Inmunológica/inmunología , Ratones , Ratones Endogámicos DBA , Linfocitos T Citotóxicos/citología , Trasplante Homólogo
19.
Proc Natl Acad Sci U S A ; 111(29): 10514-9, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-24982192

RESUMEN

Encapsulation of islets of Langerhans may represent a way to transplant islets in the absence of immunosuppression. Traditional methods for encapsulation lead to diffusional limitations imposed by the size of the capsules (600-1,000 µm in diameter), which results in core hypoxia and delayed insulin secretion in response to glucose. Moreover, the large volume of encapsulated cells does not allow implantation in sites that might be more favorable to islet cell engraftment. To address these issues, we have developed an encapsulation method that allows conformal coating of islets through microfluidics and minimizes capsule size and graft volume. In this method, capsule thickness, rather than capsule diameter, is constant and tightly defined by the microdevice geometry and the rheological properties of the immiscible fluids used for encapsulation within the microfluidic system. We have optimized the method both computationally and experimentally, and found that conformal coating allows for complete encapsulation of islets with a thin (a few tens of micrometers) continuous layer of hydrogel. Both in vitro and in vivo in syngeneic murine models of islet transplantation, the function of conformally coated islets was not compromised by encapsulation and was comparable to that of unencapsulated islets. We have further demonstrated that the structural support conferred by the coating materials protected islets from the loss of function experienced by uncoated islets during ex vivo culture.


Asunto(s)
Materiales Biocompatibles Revestidos/farmacología , Islotes Pancreáticos/efectos de los fármacos , Microfluídica/instrumentación , Alginatos/farmacología , Animales , Agregación Celular , Simulación por Computador , Diseño de Equipo , Ácido Glucurónico/farmacología , Ácidos Hexurónicos/farmacología , Hidrodinámica , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Ratones , Ratones Endogámicos C57BL , Microesferas , Modelos Biológicos , Polietilenglicoles/farmacología , Reproducibilidad de los Resultados
20.
J Exp Med ; 211(3): 441-56, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24567447

RESUMEN

Real-time imaging studies are reshaping immunological paradigms, but a visual framework is lacking for self-antigen-specific T cells at the effector phase in target tissues. To address this issue, we conducted intravital, longitudinal imaging analyses of cellular behavior in nonlymphoid target tissues to illustrate some key aspects of T cell biology. We used mouse models of T cell-mediated damage and protection of pancreatic islet grafts. Both CD4(+) and CD8(+) effector T (Teff) lymphocytes directly engaged target cells. Strikingly, juxtaposed ß cells lacking specific antigens were not subject to bystander destruction but grew substantially in days, likely by replication. In target tissue, Foxp3(+) regulatory T (Treg) cells persistently contacted Teff cells with or without involvement of CD11c(+) dendritic cells, an observation conciliating with the in vitro "trademark" of Treg function, contact-dependent suppression. This study illustrates tolerance induction by contact-based immune cell interaction in target tissues and highlights potentials of tissue regeneration under antigenic incognito in inflammatory settings.


Asunto(s)
Autoinmunidad/inmunología , Comunicación Celular/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Traslado Adoptivo , Análisis de Varianza , Animales , Autoantígenos/inmunología , Bromodesoxiuridina , Cruzamientos Genéticos , Fluorescencia , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...