Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 11(21)2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36359858

RESUMEN

Recessive Stargardt disease (STGD1) is an inherited retinopathy caused by mutations in the ABCA4 gene. The ABCA4 protein is a phospholipid-retinoid flippase in the outer segments of photoreceptors and the internal membranes of retinal pigment epithelial (RPE) cells. Here, we show that RPE cells derived via induced pluripotent stem-cell from a molecularly and clinically diagnosed STGD1 patient exhibited reduced ABCA4 protein and diminished activity compared to a normal subject. Consequently, STGD1 RPE cells accumulated intracellular autofluorescence-lipofuscin and displayed increased complement C3 activity. The level of C3 inversely correlated with the level of CD46, an early negative regulator of the complement cascade. Persistent complement dysregulation led to deposition of the membrane attack complex on the surface of RPE cells, decrease in transepithelial resistance, and subsequent cell death. These findings are strong evidence of complement-mediated RPE cell damage in STGD1, in the absence of photoreceptors, caused by reduced CD46 regulatory protein.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento , Epitelio Pigmentado de la Retina , Humanos , Enfermedad de Stargardt , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas del Sistema Complemento/metabolismo , Muerte Celular
2.
Cancer Res ; 82(20): 3763-3773, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36006995

RESUMEN

Peripheral T-cell lymphomas (PTCL) are agressive lymphomas that develop from mature T cells. The most common PTCLs are genetically, molecularly, and clinically diverse and are generally associated with dismal outcomes. While Notch signaling plays a critically important role in both the development of immature T cells and their malignant transformation, its role in PTCL is poorly understood, despite the increasingly appreciated function of Notch in regulating the proliferation and differentiation of mature T cells. Here, we demonstrate that Notch receptors and their Delta-like family ligands (DLL1/DLL4) play a pathogenic role in PTCL. Notch1 activation was observed in common PTCL subtypes, including PTCL-not otherwise specified (NOS). In a large cohort of PTCL-NOS biopsies, Notch1 activation was significantly associated with surrogate markers of proliferation. Complementary genetically engineered mouse models and spontaneous PTCL models were used to functionally examine the role of Notch signaling, and Notch1/Notch2 blockade and pan-Notch blockade using dominant-negative MAML significantly impaired the proliferation of malignant T cells and PTCL progression in these models. Treatment with DLL1/DLL4 blocking antibodies established that Notch signaling is ligand-dependent. Together, these findings reveal a role for ligand-dependent Notch signaling in driving peripheral T-cell lymphomagenesis. SIGNIFICANCE: This work demonstrates that ligand-dependent Notch activation promotes the growth and proliferation of mature T-cell lymphomas, providing new therapeutic strategies for this group of aggressive lymphomas.


Asunto(s)
Transducción de Señal , Linfocitos T , Animales , Anticuerpos Bloqueadores , Ligandos , Ratones , Receptor Notch1 , Receptores Notch/genética
3.
Cancer Res Commun ; 2(12): 1727-1737, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36970721

RESUMEN

The reciprocal relationship between malignant T cells and lymphoma-associated macrophages (LAM) within the tumor microenvironment (TME) is unique, as LAMs are well poised to provide ligands for antigen, costimulatory, and cytokine receptors that promote T-cell lymphoma growth. Conversely, malignant T cells promote the functional polarization and homeostatic survival of LAM. Therefore, we sought to determine the extent to which LAMs are a therapeutic vulnerability in these lymphomas, and to identify effective therapeutic strategies for their depletion. We utilized complementary genetically engineered mouse models and primary peripheral T-cell lymphoma (PTCL) specimens to quantify LAM expansion and proliferation. A high-throughput screen was performed to identify targeted agents that effectively deplete LAM within the context of PTCL. We observed that LAMs are dominant constituents of the TME in PTCL. Furthermore, their dominance was explained, at least in part, by their proliferation and expansion in response to PTCL-derived cytokines. Importantly, LAMs are a true dependency in these lymphomas, as their depletion significantly impaired PTCL progression. These findings were extrapolated to a large cohort of human PTCL specimens where LAM proliferation was observed. A high-throughput screen demonstrated that PTCL-derived cytokines led to relative resistance to CSF1R selective inhibitors, and culminated in the identification of dual CSF1R/JAK inhibition as a novel therapeutic strategy to deplete LAM in these aggressive lymphomas. Malignant T cells promote the expansion and proliferation of LAM, which are a bone fide dependency in these lymphomas, and are effectively depleted with a dual CSF1R/JAK inhibitor. Significance: LAMs are a therapeutic vulnerability, as their depletion impairs T-cell lymphoma disease progression. Pacritinib, a dual CSF1R/JAK inhibitor, effectively impaired LAM viability and expansion, prolonged survival in preclinical T-cell lymphoma models, and is currently being investigated as a novel therapeutic approach in these lymphomas.


Asunto(s)
Inhibidores de las Cinasas Janus , Linfoma de Células T Periférico , Linfoma de Células T , Animales , Ratones , Humanos , Linfoma de Células T Periférico/tratamiento farmacológico , Inhibidores de las Cinasas Janus/farmacología , Citocinas/farmacología , Linfoma de Células T/tratamiento farmacológico , Macrófagos , Microambiente Tumoral
4.
Diabetes ; 67(4): 769-781, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29362226

RESUMEN

Tight junctions (TJs) involve close apposition of transmembrane proteins between cells. Although TJ proteins have been studied in detail, the role of lipids is largely unknown. We addressed the role of very long-chain (VLC ≥26) ceramides in TJs using diabetes-induced loss of the blood-retinal barrier as a model. VLC fatty acids that incorporate into VLC ceramides are produced by elongase elongation of very long-chain fatty acids protein 4 (ELOVL4). ELOVL4 is significantly reduced in the diabetic retina. Overexpression of ELOVL4 significantly decreased basal permeability, inhibited vascular endothelial growth factor (VEGF)- and interleukin-1ß-induced permeability, and prevented VEGF-induced decrease in occludin expression and border staining of TJ proteins ZO-1 and claudin-5. Intravitreal delivery of AAV2-hELOVL4 reduced diabetes-induced increase in vascular permeability. Ultrastructure and lipidomic analysis revealed that ω-linked acyl-VLC ceramides colocalize with TJ complexes. Overall, normalization of retinal ELOVL4 expression could prevent blood-retinal barrier dysregulation in diabetic retinopathy through an increase in VLC ceramides and stabilization of TJs.


Asunto(s)
Barrera Hematorretinal/metabolismo , Permeabilidad Capilar/genética , Ceramidas/metabolismo , Células Endoteliales/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Vasos Retinianos/metabolismo , Uniones Estrechas/metabolismo , Animales , Bovinos , Claudina-5/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/etiología , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Células Endoteliales/ultraestructura , Humanos , Interleucina-1beta/metabolismo , Ratones , Ocludina/metabolismo , Retina/metabolismo , Vasos Retinianos/ultraestructura , Uniones Estrechas/ultraestructura , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
5.
EBioMedicine ; 22: 181-190, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28774737

RESUMEN

Diabetic retinopathy (DR) is a complication secondary to diabetes and is the number one cause of blindness among working age individuals worldwide. Despite recent therapeutic breakthroughs using pharmacotherapy, a cure for DR has yet to be realized. Several clinical trials have highlighted the vital role dyslipidemia plays in the progression of DR. Additionally, it has recently been shown that activation of Liver X receptor (LXRα/LXRß) prevents DR in diabetic animal models. LXRs are nuclear receptors that play key roles in regulating cholesterol metabolism, fatty acid metabolism and inflammation. In this manuscript, we show insight into DR pathogenesis by demonstrating an innovative signaling axis that unifies key metabolic regulators, Sirtuin 1 and LXR, in modulating retinal cholesterol metabolism and inflammation in the diabetic retina. Expression of both regulators, Sirtuin 1 and LXR, are significantly decreased in diabetic human retinal samples and in a type 2 diabetic animal model. Additionally, activation of LXR restores reverse cholesterol transport, prevents inflammation, reduces pro-inflammatory macrophages activity and prevents the formation of diabetes-induced acellular capillaries. Taken together, the work presented in this manuscript highlights the important role lipid dysregulation plays in DR progression and offers a novel potential therapeutic target for the treatment of DR.


Asunto(s)
Colesterol/metabolismo , Retinopatía Diabética/metabolismo , Receptores X del Hígado/metabolismo , Sirtuina 1/metabolismo , Animales , Bovinos , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Ratones , Retina/metabolismo , Transducción de Señal
6.
J Clin Lipidol ; 11(3): 694-703, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28457994

RESUMEN

BACKGROUND: Diabetic retinopathy is a microvascular disease that results from retinal vascular degeneration and defective repair due to diabetes-induced endothelial progenitor dysfunction. OBJECTIVE: Understanding key molecular factors involved in vascular degeneration and repair is paramount for developing effective diabetic retinopathy treatment strategies. We propose that diabetes-induced activation of acid sphingomyelinase (ASM) plays essential role in retinal endothelial and CD34+ circulating angiogenic cell (CAC) dysfunction in diabetes. METHODS: Human retinal endothelial cells (HRECs) isolated from control and diabetic donor tissue and human CD34+ CACs from control and diabetic patients were used in this study. ASM messenger RNA and protein expression were assessed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. To evaluate the effect of diabetes-induced ASM on HRECs and CD34+ CACs function, tube formation, CAC incorporation into endothelial tubes, and diurnal release of CD34+ CACs in diabetic individuals were determined. RESULTS: ASM expression level was significantly increased in HRECs isolated from diabetic compared with control donor tissue, as well as CD34+ CACs and plasma of diabetic patients. A significant decrease in tube area was observed in HRECs from diabetic donors compared with control HRECs. The tube formation deficiency was associated with increased expression of ASM in diabetic HRECs. Moreover, diabetic CD34+ CACs with high ASM showed defective incorporation into endothelial tubes. Diurnal release of CD34+ CACs was disrupted with the rhythmicity lost in diabetic patients. CONCLUSION: Collectively, these findings support that diabetes-induced ASM upregulation has a marked detrimental effect on both retinal endothelial cells and CACs.


Asunto(s)
Retinopatía Diabética/enzimología , Células Endoteliales/metabolismo , Neovascularización Patológica/patología , Retina/patología , Vasos Retinianos/fisiopatología , Esfingomielina Fosfodiesterasa/metabolismo , Anciano , Antígenos CD34/metabolismo , Ritmo Circadiano , Retinopatía Diabética/sangre , Retinopatía Diabética/patología , Retinopatía Diabética/fisiopatología , Células Endoteliales/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Vasos Retinianos/metabolismo , Esfingomielina Fosfodiesterasa/genética
7.
EBioMedicine ; 11: 138-150, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27531575

RESUMEN

Activation of pro-inflammatory and pro-angiogenic pathways in the retina and the bone marrow contributes to pathogenesis of diabetic retinopathy. We identified miR-15a as key regulator of both pro-inflammatory and pro-angiogenic pathways through direct binding and inhibition of the central enzyme in the sphingolipid metabolism, ASM, and the pro-angiogenic growth factor, VEGF-A. miR-15a was downregulated in diabetic retina and bone marrow cells. Over-expression of miR-15a downregulated, and inhibition of miR-15a upregulated ASM and VEGF-A expression in retinal cells. In addition to retinal effects, migration and retinal vascular repair function was impaired in miR-15a inhibitor-treated circulating angiogenic cells (CAC). Diabetic mice overexpressing miR-15a under Tie-2 promoter had normalized retinal permeability compared to wild type littermates. Importantly, miR-15a overexpression led to modulation toward nondiabetic levels, rather than complete inhibition of ASM and VEGF-A providing therapeutic effect without detrimental consequences of ASM and VEGF-A deficiencies.


Asunto(s)
Retinopatía Diabética/genética , Retinopatía Diabética/patología , MicroARNs/genética , Neovascularización Patológica/genética , Regiones no Traducidas 3' , Animales , Movimiento Celular/genética , Células Cultivadas , Ceramidas/biosíntesis , Diabetes Mellitus Experimental , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Interferencia de ARN , Ratas , Retina/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/metabolismo , Vasos Retinianos , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
PLoS One ; 11(1): e0146829, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26760976

RESUMEN

Diabetic retinopathy is a sight-threatening complication of diabetes, affecting 65% of patients after 10 years of the disease. Diabetic metabolic insult leads to chronic low-grade inflammation, retinal endothelial cell loss and inadequate vascular repair. This is partly due to bone marrow (BM) pathology leading to increased activity of BM-derived pro-inflammatory monocytes and impaired function of BM-derived reparative circulating angiogenic cells (CACs). We propose that diabetes has a significant long-term effect on the nature and proportion of BM-derived cells that circulate in the blood, localize to the retina and home back to their BM niche. Using a streptozotocin mouse model of diabetic retinopathy with GFP BM-transplantation, we have demonstrated that BM-derived circulating pro-inflammatory monocytes are increased in diabetes while reparative CACs are trapped in the BM and spleen, with impaired release into circulation. Diabetes also alters activation of splenocytes and BM-derived dendritic cells in response to LPS stimulation. A majority of the BM-derived GFP cells that migrate to the retina express microglial markers, while others express endothelial, pericyte and Müller cell markers. Diabetes significantly increases infiltration of BM-derived microglia in an activated state, while reducing infiltration of BM-derived endothelial progenitor cells in the retina. Further, control CACs injected into the vitreous are very efficient at migrating back to their BM niche, whereas diabetic CACs have lost this ability, indicating that the in vivo homing efficiency of diabetic CACs is dramatically decreased. Moreover, diabetes causes a significant reduction in expression of specific integrins regulating CAC migration. Collectively, these findings indicate that BM pathology in diabetes could play a role in both increased pro-inflammatory state and inadequate vascular repair contributing to diabetic retinopathy.


Asunto(s)
Células de la Médula Ósea/citología , Retinopatía Diabética/patología , Células Endoteliales/patología , Inflamación/patología , Animales , Trasplante de Médula Ósea , Recuento de Células , Quimera , Células Dendríticas/patología , Retinopatía Diabética/inmunología , Proteínas Fluorescentes Verdes/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , Microglía/patología , Microvasos/efectos de los fármacos , Microvasos/patología , Monocitos/patología , Retina/patología , Bazo/patología
9.
Stem Cells ; 34(4): 972-83, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676316

RESUMEN

The metabolic insults associated with diabetes lead to low-grade chronic inflammation, retinal endothelial cell damage, and inadequate vascular repair. This is partly due to the increased activation of bone marrow (BM)-derived proinflammatory monocytes infiltrating the retina, and the compromised function of BM-derived reparative circulating angiogenic cells (CACs), which home to sites of endothelial injury and foster vascular repair. We now propose that a metabolic link leading to activated monocytes and dysfunctional CACs in diabetes involves upregulation of a central enzyme of sphingolipid signaling, acid sphingomyelinase (ASM). Selective inhibition of ASM in the BM prevented diabetes-induced activation of BM-derived microglia-like cells and normalized proinflammatory cytokine levels in the retina. ASM upregulation in diabetic CACs caused accumulation of ceramide on their cell membrane, thereby reducing membrane fluidity and impairing CAC migration. Replacing sphingomyelin with ceramide in synthetic membrane vesicles caused a similar decrease in membrane fluidity. Inhibition of ASM in diabetic CACs improved membrane fluidity and homing of these cells to damaged retinal vessels. Collectively, these findings indicate that selective modulation of sphingolipid metabolism in BM-derived cell populations in diabetes normalizes the reparative/proinflammatory cell balance and can be explored as a novel therapeutic strategy for treating diabetic retinopathy.


Asunto(s)
Retinopatía Diabética/genética , Retinopatía Diabética/terapia , Retina/crecimiento & desarrollo , Vasos Retinianos/metabolismo , Esfingomielina Fosfodiesterasa/genética , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Ceramidas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/terapia , Ratones , Monocitos/metabolismo , Monocitos/patología , Retina/metabolismo , Retina/patología , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA