Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 2(2): e89906, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28138561

RESUMEN

Vascular abnormalities are a common component of eye diseases that often lead to vision loss. Vaso-obliteration is associated with inherited retinal degenerations, since photoreceptor atrophy lowers local metabolic demands and vascular support to those regions is no longer required. Given the degree of neurovascular crosstalk in the retina, it may be possible to use one cell type to rescue another cell type in the face of severe stress, such as hypoxia or genetically encoded cell-specific degenerations. Here, we show that intravitreally injected human endothelial colony-forming cells (ECFCs) that can be isolated and differentiated from cord blood in xeno-free media collect in the vitreous cavity and rescue vaso-obliteration and neurodegeneration in animal models of retinal disease. Furthermore, we determined that a subset of the ECFCs was more effective at anatomically and functionally preventing retinopathy; these cells expressed high levels of CD44, the hyaluronic acid receptor, and IGFBPs (insulin-like growth factor-binding proteins). Injection of cultured media from ECFCs or only recombinant human IGFBPs also rescued the ischemia phenotype. These results help us to understand the mechanism of ECFC-based therapies for ischemic insults and retinal neurodegenerative diseases.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Receptores de Hialuranos/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Isquemia/patología , Enfermedades de la Retina/patología , Neuronas Retinianas/patología , Vasos Retinianos/patología , Animales , Diferenciación Celular , Células Endoteliales/metabolismo , Células Progenitoras Endoteliales/trasplante , Sangre Fetal , Humanos , Ácido Hialurónico/metabolismo , Inyecciones Intravítreas , Ratones
2.
Sci Rep ; 6: 36659, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27834402

RESUMEN

Macrophages, key cells of the innate immune system, are known to support angiogenesis but are not believed to directly form vessel walls. Here we show that macrophages structurally form primitive, NON-ENDOTHELIAL "vessels" or vascular mimicry (VM) channels in both tumor and angiogenesis in vivo models. These channels are functionally connected to the systemic vasculature as they are perfused by intravenously injected dye. Since both models share hypoxic micro-environments, we hypothesized that hypoxia may be an important mediator of VM formation. Indeed, conditional genetic depletion of myeloid-specific HIF-1α results in decreased VM network formation, dye perfusion and tumor size. Although the macrophage VM network shares some features with an endothelial vasculature, it is ultrastructurally different. Cancer stem cells have been shown to form vascular mimicry channels. Our data demonstrates that tumor-associated macrophages also form them. The identification of this novel type of vascular mimicry may help in the development of targeted cancer therapeutics.


Asunto(s)
Vasos Sanguíneos/inmunología , Macrófagos/inmunología , Células Madre Neoplásicas/inmunología , Animales , Vasos Sanguíneos/patología , Hipoxia de la Célula/inmunología , Macrófagos/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Células Madre Neoplásicas/patología
4.
Cell Rep ; 16(3): 731-43, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27373162

RESUMEN

The effect of gut microbiota on obesity and insulin resistance is now recognized, but the underlying host-dependent mechanisms remain poorly undefined. We find that tissue inhibitor of metalloproteinase 3 knockout (Timp3(-/-)) mice fed a high-fat diet exhibit gut microbiota dysbiosis, an increase in branched chain and aromatic (BCAA) metabolites, liver steatosis, and an increase in circulating soluble IL-6 receptors (sIL6Rs). sIL6Rs can then activate inflammatory cells, such as CD11c(+) cells, which drive metabolic inflammation. Depleting the microbiota through antibiotic treatment significantly improves glucose tolerance, hepatic steatosis, and systemic inflammation, and neutralizing sIL6R signaling reduces inflammation, but only mildly impacts glucose tolerance. Collectively, our results suggest that gut microbiota is the primary driver of the observed metabolic dysfunction, which is mediated, in part, through IL-6 signaling. Our findings also identify an important role for Timp3 in mediating the effect of the microbiota in metabolic diseases.


Asunto(s)
Hígado Graso/metabolismo , Hígado Graso/patología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Microbiota/fisiología , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Disbiosis/metabolismo , Disbiosis/patología , Hígado Graso/microbiología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa/métodos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Resistencia a la Insulina/fisiología , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/microbiología , Hígado/patología , Enfermedades Metabólicas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Receptores de Interleucina-6/metabolismo , Transducción de Señal/fisiología
5.
Biochem J ; 473(4): 423-34, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621873

RESUMEN

Several forms of monogenic heritable autism spectrum disorders are associated with mutations in the neuroligin genes. The autism-linked substitution R451C in neuroligin3 induces local misfolding of its extracellular domain, causing partial retention in the ER (endoplasmic reticulum) of expressing cells. We have generated a PC12 Tet-On cell model system with inducible expression of wild-type or R451C neuroligin3 to investigate whether there is activation of the UPR (unfolded protein response) as a result of misfolded protein retention. As a positive control for protein misfolding, we also expressed the mutant G221R neuroligin3, which is known to be completely retained within the ER. Our data show that overexpression of either R451C or G221R mutant proteins leads to the activation of all three signalling branches of the UPR downstream of the stress sensors ATF6 (activating transcription factor 6), IRE1 (inositol-requiring enzyme 1) and PERK [PKR (dsRNA-dependent protein kinase)-like endoplasmic reticulum kinase]. Each branch displayed different activation profiles that partially correlated with the degree of misfolding caused by each mutation. We also show that up-regulation of BiP (immunoglobulin heavy-chain-binding protein) and CHOP [C/EBP (CCAAT/enhancer-binding protein)-homologous protein] was induced by both mutant proteins but not by wild-type neuroligin3, both in proliferative cells and cells differentiated to a neuron-like phenotype. Collectively, our data show that mutant R451C neuroligin3 activates the UPR in a novel cell model system, suggesting that this cellular response may have a role in monogenic forms of autism characterized by misfolding mutations.


Asunto(s)
Trastorno Autístico/genética , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Respuesta de Proteína Desplegada , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/química , Moléculas de Adhesión Celular Neuronal/metabolismo , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Fosforilación , Ratas , Homología de Secuencia de Aminoácido , Transcripción Genética , Regulación hacia Arriba
6.
EMBO Mol Med ; 6(5): 604-23, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24705878

RESUMEN

Current therapeutic antiangiogenic biologics used for the treatment of pathological ocular angiogenesis could have serious side effects due to their interference with normal blood vessel physiology. Here, we report the generation of novel antivascular endothelial growth factor-A (VEGF) biologics, termed VEGF "Sticky-traps," with unique properties that allow for local inhibition of angiogenesis without detectable systemic side effects. Using genetic and pharmacological approaches, we demonstrated that Sticky-traps could locally inhibit angiogenesis to at least the same extent as the original VEGF-trap that also gains whole-body access. Sticky-traps did not cause systemic effects, as shown by uncompromised wound healing and normal tracheal vessel density. Moreover, if injected intravitreally, recombinant Sticky-trap remained localized to various regions of the eye, such as the inner-limiting membrane and ciliary body, for prolonged time periods, without gaining access either to the photoreceptors/choriocapillaris area or the circulation. These unique pharmacological characteristics of Sticky-trap could allow for safe treatment of pathological angiogenesis in patients with diabetic retinopathy and retinopathy of pre-maturity.


Asunto(s)
Productos Biológicos/metabolismo , Ojo/efectos de los fármacos , Neovascularización Patológica/prevención & control , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Productos Biológicos/efectos adversos , Productos Biológicos/farmacocinética , Humanos , Receptores de Factores de Crecimiento Endotelial Vascular/efectos adversos , Receptores de Factores de Crecimiento Endotelial Vascular/farmacocinética , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Factor A de Crecimiento Endotelial Vascular/genética
7.
Diabetes ; 63(6): 2086-96, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24430438

RESUMEN

Obesity elicits immune cell infiltration of adipose tissue provoking chronic low-grade inflammation. Regulatory T cells (Tregs) are specifically reduced in adipose tissue of obese animals. Since interleukin (IL)-21 plays an important role in inducing and maintaining immune-mediated chronic inflammatory processes and negatively regulates Treg differentiation/activity, we hypothesized that it could play a role in obesity-induced insulin resistance. We found IL-21 and IL-21R mRNA expression upregulated in adipose tissue of high-fat diet (HFD) wild-type (WT) mice and in stromal vascular fraction from human obese subjects in parallel to macrophage and inflammatory markers. Interestingly, a larger infiltration of Treg cells was seen in the adipose tissue of IL-21 knockout (KO) mice compared with WT animals fed both normal diet and HFD. In a context of diet-induced obesity, IL-21 KO mice, compared with WT animals, exhibited lower body weight, improved insulin sensitivity, and decreased adipose and hepatic inflammation. This metabolic phenotype is accompanied by a higher induction of interferon regulatory factor 4 (IRF4), a transcriptional regulator of fasting lipolysis in adipose tissue. Our data suggest that IL-21 exerts negative regulation on IRF4 and Treg activity, developing and maintaining adipose tissue inflammation in the obesity state.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/inmunología , Factores Reguladores del Interferón/metabolismo , Interleucinas/metabolismo , Lipólisis , Obesidad/metabolismo , Células 3T3-L1/metabolismo , Animales , Western Blotting , Células Cultivadas , Citometría de Flujo , Inflamación/inmunología , Interleucinas/genética , Lipólisis/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/inmunología , ARN Mensajero/metabolismo , Receptores de Interleucina-21/metabolismo , Linfocitos T Reguladores , Regulación hacia Arriba
8.
J Clin Invest ; 123(11): 4900-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24084735

RESUMEN

Vascular networks develop from a growing vascular front that responds to VEGF and other guidance cues. Angiogenesis is required for normal tissue function, but, under conditions of stress, inappropriate vascularization can lead to disease. Therefore, inhibition of angiogenic sprouting may prevent neovascularization in patients with blinding neovascular eye diseases, including macular degeneration. VEGF antagonists have therapeutic benefits but also can elicit off-target effects. Here, we found that the Ras pathway, which functions downstream of a wide range of cytokines including VEGF, is active in the growing vascular front of developing and pathological vascular networks. The endogenous Ras inhibitor p120RasGAP was expressed predominately in quiescent VEGF-insensitive endothelial cells and was ectopically downregulated in multiple neovascular models. MicroRNA-132 negatively regulated p120RasGAP expression. Experimental delivery of α-miR-132 to developing mouse eyes disrupted tip cell Ras activity and prevented angiogenic sprouting. This strategy prevented ocular neovascularization in multiple rodent models even more potently than the VEGF antagonist, VEGF-trap. Targeting microRNA-132 as a therapeutic strategy may prove useful for treating multiple neovascular diseases of the eye and for preventing vision loss regardless of the neovascular stimulus.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/patología , Neovascularización Patológica/prevención & control , Proteínas ras/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Fisiológica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Neovascularización Retiniana/prevención & control , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Proteínas ras/metabolismo
9.
EMBO Mol Med ; 5(3): 441-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23401241

RESUMEN

ADAM17 and its inhibitor TIMP3 are involved in nephropathy, but their role in diabetic kidney disease (DKD) is unclear. Diabetic Timp3(-/-) mice showed increased albuminuria, increased membrane thickness and mesangial expansion. Microarray profiling uncovered a significant reduction of Foxo1 expression in diabetic Timp3(-/-) mice compared to WT, along with FoxO1 target genes involved in autophagy, while STAT1, a repressor of FoxO1 transcription, was increased. Re-expression of Timp3 in Timp3(-/-) mesangial cells rescued the expression of Foxo1 and its targets, and decreased STAT1 expression to control levels; abolishing STAT1 expression led to a rescue of FoxO1, evoking a role of STAT1 in linking Timp3 deficiency to FoxO1. Studies on kidney biopsies from patients with diabetic nephropathy confirmed a significant reduction in TIMP3, FoxO1 and FoxO1 target genes involved in autophagy compared to controls, while STAT1 expression was strongly increased. Our study suggests that loss of TIMP3 is a hallmark of DKD in human and mouse models and designates TIMP3 as a new possible therapeutic target for diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , Factores de Transcripción Forkhead/metabolismo , Glomérulos Renales/metabolismo , Factor de Transcripción STAT1/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/deficiencia , Albuminuria/etiología , Albuminuria/metabolismo , Animales , Autofagia , Biopsia , Línea Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Glomérulos Renales/patología , Células Mesangiales/metabolismo , Ratones , Ratones Noqueados , Cultivo Primario de Células , Interferencia de ARN , Factor de Transcripción STAT1/genética , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-3/genética , Transfección
10.
Int J Inflam ; 2012: 360147, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701810

RESUMEN

Cardiovascular disease is the leading cause of death in the general population; traditional risk factors seem inadequate to explain completely the remarkable prevalence of cardiovascular mortality and morbidity observed in the uremic population. A role for chronic inflammation has been well established in the development of atherosclerotic disease, and, on the basis of these observations, atherosclerosis might be considered an inflammatory disease. Inflammation has been implicated in the etiology of coronary artery disease in the general population, and traditional inflammatory biomarkers such as C-reactive protein (CRP) and interleukin-6 (IL-6) have been shown to predict cardiovascular events in both symptomatic and asymptomatic individuals as well as those in the uremic population. Later on, new nontraditional markers were related to the risk of cardiovascular morbidity and mortality in general and in uremic population. As a consequence of the expanding research base and availability of assays, the number of inflammatory marker tests ordered by clinicians for cardiovascular disease (CVD) risk prediction has grown rapidly and several commercial assays have become available. So, up to now we can consider that several new nontraditional markers as CD40-CD40 ligand system and pentraxin-3 seem to be significant features of cardiovascular disease in general and in ESRD population.

11.
Arterioscler Thromb Vasc Biol ; 32(1): 74-81, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22015660

RESUMEN

OBJECTIVE: Tissue inhibitor of metalloproteinase 3 (TIMP3) is a stromal protein that inhibits the activity of proteases and receptors. TIMP3 is downregulated in metabolic and inflammatory disorders, such as type 2 diabetes mellitus and atherosclerosis, particularly in regions enriched with monocyte/macrophage cells. To investigate the role of TIMP3 in atherosclerosis, we generated a new mouse model in which Timp3 was overexpressed in the atherosclerotic plaque via a macrophage-specific promoter (MacT3). We elucidated any potential antiatherosclerotic effects of TIMP3, including regulation of monocyte/macrophage recruitment within atherosclerotic plaques, in MacT3 mice crossbred with low-density lipoprotein receptor knockout (LDLR(-/-)) mice. METHODS AND RESULTS: MacT3/LDLR(-/-) mice had an improvement of atherosclerosis and metabolic parameters compared with LDLR(-/-). En face aorta and aortic root examination of MacT3/LDLR(-/-) mice revealed smaller atherosclerotic plaques with features of stability, such as increased collagen content and decreased necrotic core formation. Atherosclerotic plaques in MacT3/LDLR(-/-) mice contained fewer T cells and macrophages. Furthermore, TIMP3 overexpression in macrophages resulted in reduced oxidative stress signals, as evidenced by lower lipid peroxidation, protein carbonylation, and nitration in atheromas. CONCLUSIONS: Our study confirmed that macrophage-specific overexpression of TIMP3 decreases the inflammatory content and the amplitude of atherosclerotic plaques in mice.


Asunto(s)
Aterosclerosis/prevención & control , Macrófagos/metabolismo , Receptores de LDL/deficiencia , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Dieta Aterogénica/efectos adversos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores de LDL/genética , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba
13.
Nephrol Dial Transplant ; 26(8): 2641-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21325348

RESUMEN

BACKGROUND: Resistance to erythropoiesis-stimulating agents (ESAs) is often associated with chronic inflammation. Here, we investigated how anaemia, ESA resistance and the plasma levels of biological markers of inflammation could influence all-cause and cardiovascular disease morbidity and mortality. METHODS: Seven hundred and fifty-three haemodialysis (HD) patients (mean age 66 ± 14.2 years, mean dialytic age 70 ± 77 months and diabetes 18.8%) were enrolled and followed-up for 36 months. Demographic, clinical and laboratory data, co-morbidity conditions, administered drugs, all-cause mortality and fatal/non-fatal cardiovascular (CV) events were recorded. We measured ESA resistance index, C-reactive protein (CRP) and interleukin-6 (IL-6). RESULTS: Six hundred and fifty-one patients (86.4%) received ESAs. Patients with haemoglobin level <11 g/dL (n = 225) showed increased risk of CV [relative risk (RR) 1.415, 95% confidence interval (CI) 1.046-1.914] and overall mortality (RR 1.897, 95% CI 1.423-2.530) versus patients with haemoglobin levels >11 g/dL. ESA resistance values categorized into quartiles (Quartile I <5.6, Quartile II 5.7-9.6, Quartile III 9.7-15.4 and Quartile IV >15.4) correlated with all-cause mortality and fatal/non-fatal CV events (RR 1.97, 95% CI 1.392-2.786; RR 1.619, 95% CI 1.123-2.332, respectively). Furthermore, albumin was significantly reduced versus reference patients and correlated with all-cause mortality and CV events; CRP levels were higher in hyporesponders (Quartile IV) (P < 0.001) and predicted all-cause mortality and CV events. IL-6 but not CRP was a strong predictor of ESA resistance. CONCLUSIONS: ESA responsiveness can be considered a strong prognostic factor in HD patients and seems to be tightly related to protein-energy wasting and inflammation.


Asunto(s)
Anemia/complicaciones , Anemia/tratamiento farmacológico , Resistencia a Medicamentos , Hematínicos/efectos adversos , Inflamación/etiología , Fallo Renal Crónico/mortalidad , Diálisis Renal/efectos adversos , Anciano , Anemia/mortalidad , Biomarcadores/metabolismo , Proteína C-Reactiva/metabolismo , Femenino , Estudios de Seguimiento , Tasa de Filtración Glomerular , Humanos , Inflamación/mortalidad , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Pruebas de Función Renal , Masculino , Pronóstico , Diálisis Renal/métodos , Tasa de Supervivencia
14.
Sci Rep ; 1: 76, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355595

RESUMEN

Diabetic retinopathy is the leading cause of visual loss in individuals under the age of 55. Umbilical cord blood (UCB)-derived myeloid progenitor cells have been shown to decrease neuronal damage associated with ischemia in the central nervous system. In this study we show that UCB-derived CD14(+) progenitor cells provide rescue effects in a mouse model of ischemic retinopathy by promoting physiological angiogenesis and reducing associated inflammation. We use confocal microscopy to trace the fate of injected human UCB-derived CD14(+) cells and PCR with species-specific probes to investigate their gene expression profile before and after injection. Metabolomic analysis measures changes induced by CD14(+) cells. Our results demonstrate that human cells differentiate in vivo into M2 macrophages and induce the polarization of resident M2 macrophages. This leads to stabilization of the ischemia-injured retinal vasculature by modulating the inflammatory response, reducing oxidative stress and apoptosis and promoting tissue repair.


Asunto(s)
Modelos Animales de Enfermedad , Isquemia/patología , Macrófagos/fisiología , Enfermedades de la Retina/patología , Vasos Retinianos/patología , Animales , Células Cultivadas , Humanos , Receptores de Lipopolisacáridos/inmunología , Macrófagos/inmunología , Ratones , Microscopía Confocal , Reacción en Cadena de la Polimerasa
15.
J Clin Invest ; 120(9): 3012-21, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20811157

RESUMEN

Dramatic advances in the field of stem cell research have raised the possibility of using these cells to treat a variety of diseases. The eye is an excellent target organ for such cell-based therapeutics due to its ready accessibility, the prevalence of vasculo- and neurodegenerative diseases affecting vision, and the availability of animal models to demonstrate proof of concept. In fact, stem cell therapies have already been applied to the treatment of disease affecting the ocular surface, leading to preservation of vision. Diseases in the back of the eye, such as macular degeneration, diabetic retinopathy, and inherited retinal degenerations, present greater challenges, but rapidly emerging stem cell technologies hold the promise of autologous grafts to stabilize vision loss through cellular replacement or paracrine rescue effects.


Asunto(s)
Degeneración Macular/terapia , Degeneración Retiniana/terapia , Trasplante de Células Madre/métodos , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Ojo/anatomía & histología , Ojo/patología , Humanos
16.
J Nephrol ; 23(5): 556-62, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20349412

RESUMEN

BACKGROUND: Despite substantial progress in medical care, the mortality rate remains unacceptably high in dialysis patients. Evidence suggests that bone mineral dismetabolism (CKD-MBD) might contribute to this burden of death. However, to date only a few papers have investigated the clinical relevance of serum mineral derangements and the impact of different therapeutic strategies on mortality in a homogeneous cohort of south European dialysis patients. METHODS: The RISCAVID study was a prospective, observational study in which all patients receiving hemodialysis (HD) in the north-western region of Toscany in June 2004 were enrolled (N=757) and followed up for 24 months. RESULTS: At study entry, only 71 (9%) patients of the entire study cohort exhibited an optimal control of serum phosphorous (Pi), calcium (Ca), calciumX-phosphorous product (CAXPi) and intact parathyroidhormone (iPTH) according to the Kidney Disease Outcomes Quality Initiative (K/DOQI) clinical guidelines. Despite a similar prevalence, the severity of CKD-MBD appeared different to the results reported in the USA. Interestingly, none of the serum biomarkers or number of serum biomarkers within KDOQI targets was independently associated with all-cause and cardiovascular (CV) mortality. Among treatments, Sevelamer was the only drug independently associated with lower all-cause and cardiovascular mortality (p<0.001). CONCLUSION: The RISCAVID study highlights the difficulty of controlling bone mineral metabolism in HD patients and lends support to the hypothesis that a carefully chosen phosphate binder might impact survival in HD patients.


Asunto(s)
Calcio/sangre , Hormona Paratiroidea/sangre , Fosfatos/sangre , Diálisis Renal/mortalidad , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Poliaminas/uso terapéutico , Estudios Prospectivos , Sevelamer
17.
Methods Enzymol ; 444: 115-58, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19007663

RESUMEN

During normal retinal vascular development, vascular endothelial cells proliferate and migrate through the extracellular matrix in response to a variety of cytokines, leading to the formation of new blood vessels in a highly ordered fashion. However, abnormal angiogenesis contributes to the vast majority of diseases that cause catastrophic loss of vision. During abnormal neovascularization of the iris, retina, or choroid, angiogenesis is unregulated and usually results in the formation of dysfunctional blood vessels. Multiple models of ocular angiogenesis exist which recapitulate particular aspects of both normal and pathological neovascularization. These experimental methods are useful for studying the mechanisms of normal developmental angiogenesis, as well as studying various aspects of pathological angiogenesis including ischemic retinopathies, vascular leak, and choroidal neovascularization. This chapter will outline several protocols used to study ocular angiogenesis, put the protocols into brief historical context, and describe some of the questions for which these protocols are commonly used.


Asunto(s)
Ojo/irrigación sanguínea , Modelos Biológicos , Neovascularización Fisiológica , Animales , Oftalmopatías/inducido químicamente , Ratones , Procedimientos Quirúrgicos Oftalmológicos , Oxígeno/farmacología
18.
Angiogenesis ; 10(2): 89-101, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17372851

RESUMEN

Nothing more dramatically captures the imagination of the visually impaired patient or the ophthalmologist treating them than the possibility of rebuilding a damaged retina or vasculature with "stem cells." Stem cells (SC) have been isolated from adult tissues and represent a pool of cells that may serve to facilitate rescue/repair of damaged tissue following injury or stress. We propose a new paradigm to "mature" otherwise immature neovasculature or, better yet, stabilize existing vasculature to hypoxic damage. This may be possible through the use of autologous bone marrow (BM) or cord blood derived hematopoietic SC that selectively target sites of neovascularization and gliosis where they provide vasculo- and neurotrophic effects. We have demonstrated that adult BM contains a population of endothelial and myeloid progenitor cells that can target activated astrocytes, a hallmark of many ocular diseases, and participate in normal developmental, or injury-induced, angiogenesis in the adult. Intravitreal injection of these cells from mice and humans can prevent retinal vascular degeneration ordinarily observed in mouse models of retinal degeneration; this vascular rescue correlates with functional neuronal rescue as well. The use of autologous adult BM derived SC grafts for the treatment of retinal vascular and degenerative diseases represents a novel conceptual approach that may make it possible to "mature" otherwise immature neovasculature, stabilize existing vasculature to hypoxic damage and/or rescue and protect retinal neurons from undergoing apoptosis. Such a therapeutic approach would obviate the need to employ destructive treatment modalities and would facilitate vascularization of ischemic and otherwise damaged retinal tissue.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neovascularización Fisiológica , Enfermedades de la Retina/fisiopatología , Enfermedades de la Retina/terapia , Vasos Retinianos/fisiología , Trasplante de Células Madre , Adulto , Células de la Médula Ósea/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Endotelio Vascular/citología , Humanos , Vasos Retinianos/fisiopatología , Seguridad
19.
Diabetes ; 55(8): 2231-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873685

RESUMEN

Dysfunction of mature endothelial cells is thought to play a major role in both micro- and macrovascular complications of diabetes. However, recent advances in biology of endothelial progenitor cells (EPCs) have highlighted their involvement in diabetes complications. To determine the effect of glucotoxicity on EPCs, human EPCs have been isolated from peripheral blood mononuclear cells of healthy donors and cultured in the presence or absence of high glucose (33 mmol/l) or high glucose plus benfotiamine to scavenge glucotoxicity. Morphological analysis revealed that high glucose significantly affected the number of endothelial cell colony forming units, uptake and binding of acLDL and Lectin-1, and the ability to differentiate into CD31- and vascular endothelial growth factor receptor 2-positive cells. Functional analysis outlined a reduced EPC involvement in de novo tube formation, when cocultured with mature endothelial cells (human umbilical vein endothelial cells) on matrigel. To explain the observed phenotypes, we have investigated the signal transduction pathways known to be involved in EPC growth and differentiation. Our results indicate that hyperglycemia impairs EPC differentiation and that the process can be restored by benfotiamine administration, via the modulation of Akt/FoxO1 activity.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Factores de Transcripción Forkhead/fisiología , Glucosa/toxicidad , Proteínas Proto-Oncogénicas c-akt/fisiología , Células Madre/efectos de los fármacos , Tiamina/análogos & derivados , Androstadienos/farmacología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Factores de Transcripción Forkhead/efectos de los fármacos , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Tiamina/farmacología , Wortmanina
20.
J Clin Invest ; 115(12): 3494-505, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16294222

RESUMEN

Activation of inflammatory pathways may contribute to the beginning and the progression of both atherosclerosis and type 2 diabetes. Here we report a novel interaction between insulin action and control of inflammation, resulting in glucose intolerance and vascular inflammation and amenable to therapeutic modulation. In insulin receptor heterozygous (Insr+/-) mice, we identified the deficiency of tissue inhibitor of metalloproteinase 3 (Timp3, an inhibitor of both TNF-alpha-converting enzyme [TACE] and MMPs) as a common bond between glucose intolerance and vascular inflammation. Among Insr+/- mice, those that develop diabetes have reduced Timp3 and increased TACE activity. Unchecked TACE activity causes an increase in levels of soluble TNF-alpha, which subsequently promotes diabetes and vascular inflammation. Double heterozygous Insr+/-Timp3+/- mice develop mild hyperglycemia and hyperinsulinemia at 3 months and overt glucose intolerance and hyperinsulinemia at 6 months. A therapeutic role for Timp3/TACE modulation is supported by the observation that pharmacological inhibition of TACE led to marked reduction of hyperglycemia and vascular inflammation in Insr+/- diabetic mice, as well as by the observation of increased insulin sensitivity in Tace+/- mice compared with WT mice. Our results suggest that an interplay between reduced insulin action and unchecked TACE activity promotes diabetes and vascular inflammation.


Asunto(s)
Inflamación/genética , Receptor de Insulina/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/fisiología , Factor de Necrosis Tumoral alfa/genética , Análisis de Varianza , Animales , Desoxiglucosa/metabolismo , Diabetes Mellitus/patología , Electroforesis en Gel de Poliacrilamida , Perfilación de la Expresión Génica , Vectores Genéticos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucógeno/química , Heterocigoto , Homeostasis , Hiperglucemia/patología , Hiperinsulinismo/patología , Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/patología , Músculos/patología , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , ARN/química , ARN/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...