Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Stem Cells Int ; 2017: 9216502, 2017.
Article in English | MEDLINE | ID: mdl-29527228

ABSTRACT

Obesity is associated with enhanced tumor growth and progression. Within the adipose tissue are adipose-derived stromal/stem cells (ASCs) that have been shown to convert into carcinoma-associated fibroblast (CAFs) in the presence of tumor-derived factors. However, the impact of obesity on the ASCs and on the conversion of ASCs into CAFs has not been demonstrated. In the current study, ASCs isolated from lean donors (BMI < 25; lnASCs) were compared with ASCs isolated from obese donors (BMI > 30, obASCs). The contribution of tumor-derived factors on the conversion of ASCs to CAFs was investigated. Following exposure to cancer cells, obASCs expressed higher levels of CAF markers, including NG2, alpha-SMA, VEGF, FAP, and FSP, compared to lnASCs. To investigate the crosstalk between ASCs and breast cancer cells, MCF7 cells were serially cocultured with lnASCs or obASCs. After coculture with lnASCs and obASCs, MCF7 cells demonstrated enhanced proliferation and expressed an invasive phenotype morphologically, with more pronounced effects following exposure to obASCs. Long-term exposure to obASCs also enhanced the expression of protumorgenic factors. Together, these results suggest that obesity alters ASCs to favor their rapid conversion into CAFs, which in turn enhances the proliferative rate, the phenotype, and gene expression profile of breast cancer cells.

3.
Nat Med ; 22(4): 439-45, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26974308

ABSTRACT

Tissues with high metabolic rates often use lipids, as well as glucose, for energy, conferring a survival advantage during feast and famine. Current dogma suggests that high-energy-consuming photoreceptors depend on glucose. Here we show that the retina also uses fatty acid ß-oxidation for energy. Moreover, we identify a lipid sensor, free fatty acid receptor 1 (Ffar1), that curbs glucose uptake when fatty acids are available. Very-low-density lipoprotein receptor (Vldlr), which is present in photoreceptors and is expressed in other tissues with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acid. In the retinas of Vldlr(-/-) mice with low fatty acid uptake but high circulating lipid levels, we found that Ffar1 suppresses expression of the glucose transporter Glut1. Impaired glucose entry into photoreceptors results in a dual (lipid and glucose) fuel shortage and a reduction in the levels of the Krebs cycle intermediate α-ketoglutarate (α-KG). Low α-KG levels promotes stabilization of hypoxia-induced factor 1a (Hif1a) and secretion of vascular endothelial growth factor A (Vegfa) by starved Vldlr(-/-) photoreceptors, leading to neovascularization. The aberrant vessels in the Vldlr(-/-) retinas, which invade normally avascular photoreceptors, are reminiscent of the vascular defects in retinal angiomatous proliferation, a subset of neovascular age-related macular degeneration (AMD), which is associated with high vitreous VEGFA levels in humans. Dysregulated lipid and glucose photoreceptor energy metabolism may therefore be a driving force in macular telangiectasia, neovascular AMD and other retinal diseases.


Subject(s)
Fatty Acids/metabolism , Macular Degeneration/metabolism , Photoreceptor Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, LDL/metabolism , Retina/metabolism , Animals , Gene Expression Regulation , Glucose/metabolism , Humans , Ketoglutaric Acids/metabolism , Lipid Metabolism/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Oxidation-Reduction , Photoreceptor Cells/pathology , Receptors, G-Protein-Coupled/biosynthesis , Receptors, LDL/genetics , Retina/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Am J Clin Nutr ; 101(4): 879-88, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25833984

ABSTRACT

BACKGROUND: Retinopathy of prematurity (ROP) is a vision-threatening disease in premature infants. Serum adiponectin (APN) concentrations positively correlate with postnatal growth and gestational age, important risk factors for ROP development. Dietary ω-3 (n-3) long-chain polyunsaturated fatty acids (ω-3 LCPUFAs) suppress ROP and oxygen-induced retinopathy (OIR) in a mouse model of human ROP, but the mechanism is not fully understood. OBJECTIVE: We examined the role of APN in ROP development and whether circulating APN concentrations are increased by dietary ω-3 LCPUFAs to mediate the protective effect in ROP. DESIGN: Serum APN concentrations were correlated with ROP development and serum ω-3 LCPUFA concentrations in preterm infants. Mouse OIR was then used to determine whether ω-3 LCPUFA supplementation increases serum APN concentrations, which then suppress retinopathy. RESULTS: We found that in preterm infants, low serum APN concentrations positively correlate with ROP, and serum APN concentrations positively correlate with serum ω-3 LCPUFA concentrations. In mouse OIR, serum total APN and bioactive high-molecular-weight APN concentrations are increased by ω-3 LCPUFA feed. White adipose tissue, where APN is produced and assembled in the endoplasmic reticulum, is the major source of serum APN. In mouse OIR, adipose endoplasmic reticulum stress is increased, and APN production is suppressed. ω-3 LCPUFA feed in mice increases APN production by reducing adipose endoplasmic reticulum stress markers. Dietary ω-3 LCPUFA suppression of neovascularization is reduced from 70% to 10% with APN deficiency. APN receptors localize in the retina, particularly to pathologic neovessels. CONCLUSION: Our findings suggest that increasing APN by ω-3 LCPUFA supplementation in total parental nutrition for preterm infants may suppress ROP.


Subject(s)
Adiponectin/blood , Adiposity/drug effects , Endoplasmic Reticulum Stress/drug effects , Fatty Acids, Omega-3/administration & dosage , Retinal Neovascularization/drug therapy , 3T3-L1 Cells , Adiponectin/deficiency , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Animals, Newborn/blood , Disease Models, Animal , Fatty Acids, Omega-3/blood , Female , Humans , Infant, Newborn , Infant, Premature/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies , Retina/drug effects , Retina/metabolism , Retinal Neovascularization/blood , Retinopathy of Prematurity/blood , Retinopathy of Prematurity/drug therapy
5.
Invest Ophthalmol Vis Sci ; 55(12): 8267-77, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25414194

ABSTRACT

PURPOSE: Pathological neovessel formation impacts many blinding vascular eye diseases. Identification of molecular signatures distinguishing pathological neovascularization from normal quiescent vessels is critical for developing new interventions. Twist-related protein 1 (TWIST1) is a transcription factor important in tumor and pulmonary angiogenesis. This study investigated the potential role of TWIST1 in modulating pathological ocular angiogenesis in mice. METHODS: Twist1 expression and localization were analyzed in a mouse model of oxygen-induced retinopathy (OIR). Pathological ocular angiogenesis in Tie2-driven conditional Twist1 knockout mice were evaluated in both OIR and laser-induced choroidal neovascularization models. In addition, the effects of TWIST1 on angiogenesis and endothelial cell function were analyzed in sprouting assays of aortic rings and choroidal explants isolated from Twist1 knockout mice, and in human retinal microvascular endothelial cells treated with TWIST1 small interfering RNA (siRNA). RESULTS: TWIST1 is highly enriched in pathological neovessels in OIR retinas. Conditional Tie2-driven depletion of Twist1 significantly suppressed pathological neovessels in OIR without impacting developmental retinal angiogenesis. In a laser-induced choroidal neovascularization model, Twist1 deficiency also resulted in significantly smaller lesions with decreased vascular leakage. In addition, loss of Twist1 significantly decreased vascular sprouting in both aortic ring and choroid explants. Knockdown of TWIST1 in endothelial cells led to dampened expression of vascular endothelial growth factor receptor 2 (VEGFR2) and decreased endothelial cell proliferation. CONCLUSIONS: Our study suggests that TWIST1 is a novel regulator of pathologic ocular angiogenesis and may represent a new molecular target for developing potential therapeutic treatments to suppress pathological neovascularization in vascular eye diseases.


Subject(s)
Choroidal Neovascularization/physiopathology , Nuclear Proteins/physiology , Retinal Neovascularization/physiopathology , Twist-Related Protein 1/physiology , Animals , Choroid/blood supply , Disease Models, Animal , Endothelial Cells/physiology , Fluorescein Angiography , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Neovascularization, Pathologic/physiopathology , Nuclear Proteins/deficiency , Oxygen/pharmacology , RNA, Messenger/metabolism , Retinal Vessels/cytology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Twist-Related Protein 1/deficiency , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
PLoS One ; 9(1): e85031, 2014.
Article in English | MEDLINE | ID: mdl-24416337

ABSTRACT

Proliferative retinopathy is a leading cause of blindness, including retinopathy of prematurity (ROP) in children and diabetic retinopathy in adults. Retinopathy is characterized by an initial phase of vessel loss, leading to tissue ischemia and hypoxia, followed by sight threatening pathologic neovascularization in the second phase. Previously we found that Sirtuin1 (Sirt1), a metabolically dependent protein deacetylase, regulates vascular regeneration in a mouse model of oxygen-induced proliferative retinopathy (OIR), as neuronal depletion of Sirt1 in retina worsens retinopathy. In this study we assessed whether over-expression of Sirtuin1 in retinal neurons and vessels achieved by crossing Sirt1 over-expressing flox mice with Nestin-Cre mice or Tie2-Cre mice, respectively, may protect against retinopathy. We found that over-expression of Sirt1 in Nestin expressing retinal neurons does not impact vaso-obliteration or pathologic neovascularization in OIR, nor does it influence neuronal degeneration in OIR. Similarly, increased expression of Sirt1 in Tie2 expressing vascular endothelial cells and monocytes/macrophages does not protect retinal vessels in OIR. In addition to the genetic approaches, dietary supplement with Sirt1 activators, resveratrol or SRT1720, were fed to wild type mice with OIR. Neither treatment showed significant vaso-protective effects in retinopathy. Together these results indicate that although endogenous Sirt1 is important as a stress-induced protector in retinopathy, over-expression of Sirt1 or treatment with small molecule activators at the examined doses do not provide additional protection against retinopathy in mice. Further studies are needed to examine in depth whether increasing levels of Sirt1 may serve as a potential therapeutic approach to treat or prevent retinopathy.


Subject(s)
Nerve Degeneration/genetics , Neurons/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Sirtuin 1/genetics , Animals , Crosses, Genetic , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Integrases/genetics , Integrases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Neovascularization, Pathologic , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nestin/genetics , Nestin/metabolism , Neurons/drug effects , Neurons/pathology , Oxygen/adverse effects , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Resveratrol , Retina/drug effects , Retina/pathology , Retinal Degeneration/chemically induced , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Sirtuin 1/metabolism , Stilbenes/pharmacology
7.
Angiogenesis ; 16(4): 985-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23912262

ABSTRACT

Regeneration of blood vessels in ischemic neuronal tissue is critical to reduce tissue damage in diseases. In proliferative retinopathy, initial vessel loss leads to retinal ischemia, which can induce either regrowth of vessels to restore normal metabolism and minimize damage, or progress to hypoxia-induced sight-threatening pathologic vaso-proliferation. It is not well understood how retinal neurons mediate regeneration of vascular growth in response to ischemic insults. In this study we aim to investigate the potential role of Sirtuin 1 (Sirt1), a metabolically-regulated protein deacetylase, in mediating the response of ischemic neurons to regulate vascular regrowth in a mouse model of oxygen-induced ischemic retinopathy (OIR). We found that Sirt1 is highly induced in the avascular ischemic retina in OIR. Conditional depletion of neuronal Sirt1 leads to significantly decreased retinal vascular regeneration into the avascular zone and increased hypoxia-induced pathologic vascular growth. This effect is likely independent of PGC-1α, a known Sirt1 target, as absence of PGC-1α in knockout mice does not impact vascular growth in retinopathy. We found that neuronal Sirt1 controls vascular regrowth in part through modulating deacetylation and stability of hypoxia-induced factor 1α and 2α, and thereby modulating expression of angiogenic factors. These results indicate that ischemic neurons induce Sirt1 to promote revascularization into ischemic neuronal areas, suggesting a novel role of neuronal Sirt1 in mediating vascular regeneration in ischemic conditions, with potential implications beyond retinopathy.


Subject(s)
Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Neurons/metabolism , Regeneration/physiology , Retinal Vessels/physiology , Retinopathy of Prematurity , Sirtuin 1/physiology , Angiogenic Proteins/biosynthesis , Angiogenic Proteins/genetics , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbazoles/pharmacology , Cell Line , Disease Models, Animal , Ischemia/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Oxygen/toxicity , Oxygen Inhalation Therapy/adverse effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/enzymology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/deficiency , Sirtuin 1/genetics , Transcription Factors/biosynthesis , Transcription Factors/deficiency , Transcription Factors/genetics , Up-Regulation
8.
PLoS One ; 8(7): e69552, 2013.
Article in English | MEDLINE | ID: mdl-23922736

ABSTRACT

Angiogenesis of the microvasculature is central to the etiology of many diseases including proliferative retinopathy, age-related macular degeneration and cancer. A mouse model of microvascular angiogenesis would be very valuable and enable access to a wide range of genetically manipulated tissues that closely approximate small blood vessel growth in vivo. Vascular endothelial cells cultured in vitro are widely used, however, isolating pure vascular murine endothelial cells is technically challenging. A microvascular mouse explant model that is robust, quantitative and can be reproduced without difficulty would overcome these limitations. Here we characterized and optimized for reproducibility an organotypic microvascular angiogenesis mouse and rat model from the choroid, a microvascular bed in the posterior of eye. The choroidal tissues from C57BL/6J and 129S6/SvEvTac mice and Sprague Dawley rats were isolated and incubated in Matrigel. Vascular sprouting was comparable between choroid samples obtained from different animals of the same genetic background. The sprouting area, normalized to controls, was highly reproducible between independent experiments. We developed a semi-automated macro in ImageJ software to allow for more efficient quantification of sprouting area. Isolated choroid explants responded to manipulation of the external environment while maintaining the local interactions of endothelial cells with neighboring cells, including pericytes and macrophages as evidenced by immunohistochemistry and fluorescence-activated cell sorting (FACS) analysis. This reproducible ex vivo angiogenesis assay can be used to evaluate angiogenic potential of pharmacologic compounds on microvessels and can take advantage of genetically manipulated mouse tissue for microvascular disease research.


Subject(s)
Biological Assay/methods , Choroid/blood supply , Microvessels/physiology , Models, Biological , Neovascularization, Physiologic , Aging/physiology , Angiogenesis Inducing Agents/pharmacology , Animals , Biological Assay/standards , Choroid/drug effects , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Monocytes/cytology , Monocytes/metabolism , Neovascularization, Physiologic/drug effects , Pericytes/cytology , Pericytes/metabolism , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Retinal Pigment Epithelium/physiology
9.
Blood ; 120(14): 2925-9, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-22791286

ABSTRACT

Inflammatory cytokines and growth factors drive angiogenesis independently; however, their integrated role in pathologic and physiologic angiogenesis is not fully understood. Suppressor of cytokine signaling-3 (SOCS3) is an inducible negative feedback regulator of inflammation and growth factor signaling. In the present study, we show that SOCS3 curbs pathologic angiogenesis. Using a Cre/Lox system, we deleted SOCS3 in vessels and studied developmental and pathologic angiogenesis in murine models of oxygen-induced retinopathy and cancer. Conditional loss of SOCS3 leads to increased pathologic neovascularization, resulting in pronounced retinopathy and increased tumor size. In contrast, physiologic vascularization is not regulated by SOCS3. In vitro, SOCS3 knockdown increases proliferation and sprouting of endothelial cells costimulated with IGF-1 and TNFα via reduced feedback inhibition of the STAT3 and mTOR pathways. These results identify SOCS3 as a pivotal endogenous feedback inhibitor of pathologic angiogenesis and a potential therapeutic target acting at the converging crossroads of growth factor- and cytokine-induced vessel growth.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Hypoxia/pathology , Melanoma, Experimental/prevention & control , Neovascularization, Pathologic/prevention & control , Paraneoplastic Syndromes, Ocular/prevention & control , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Blotting, Western , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Integrases/metabolism , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/etiology , Paraneoplastic Syndromes, Ocular/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Invest Ophthalmol Vis Sci ; 53(6): 2968-77, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22491401

ABSTRACT

PURPOSE: Retinopathy of prematurity (ROP) is a leading cause of blindness in children and is, in its most severe form, characterized by uncontrolled growth of vision-threatening pathologic vessels. Propranolol, a nonselective ß-adrenergic receptor blocker, was reported to protect against pathologic retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). Based on this single animal study using nonstandard evaluation of retinopathy, clinical trials are currently ongoing to evaluate propranolol treatment in stage 2 ROP patients who tend to experience spontaneous disease regression and are at low risk of blindness. Because these ROP patients are vulnerable premature infants who are still in a fragile state of incomplete development, the efficacy of propranolol treatment in retinopathy needs to be evaluated thoroughly in preclinical animal models of retinopathy and potential benefits weighed against potential adverse effects. METHODS: Retinopathy was induced by exposing neonatal mice to 75% oxygen from postnatal day (P) 7 to P12. Three routes of propranolol treatment were assessed from P12 to P16: oral gavage, intraperitoneal injection, or subcutaneous injection, with doses varying between 2 and 60 mg/kg/day. At P17, retinal flatmounts were stained with isolectin and quantified with a standard protocol to measure vasoobliteration and pathologic neovascularization. Retinal gene expression was analyzed with qRT-PCR using RNA isolated from retinas of control and propranolol-treated pups. RESULTS: None of the treatment approaches at any dose of propranolol (up to 60 mg/kg/day) were effective in preventing the development of retinopathy in a mouse model of OIR, evaluated using standard techniques. Propranolol treatment also did not change retinal expression of angiogenic factors including vascular endothelial growth factor. CONCLUSIONS: Propranolol treatment via three routes and up to 30 times the standard human dose failed to suppress retinopathy development in mice. These data bring into question whether propranolol through inhibition of ß-adrenergic receptors is an appropriate therapeutic approach for treating ROP.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Propranolol/administration & dosage , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/prevention & control , Administration, Oral , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Animals, Newborn , Cell Proliferation , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Erythropoietin/genetics , Erythropoietin/metabolism , Gene Expression Profiling , Humans , In Vitro Techniques , Infant, Newborn , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Oxygen/toxicity , RNA/metabolism , Receptors, Adrenergic, beta/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Retina/cytology , Retinal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...