Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Front Cell Dev Biol ; 9: 631413, 2021.
Article En | MEDLINE | ID: mdl-34222224

Retinal pigment epithelium (RPE), a postmitotic monolayer located between the neuroretina and choroid, supports the retina and is closely associated with vision loss diseases such as age-related macular degeneration (AMD) upon dysfunction. Although environmental stresses are known to play critical roles in AMD pathogenesis and the roles of other stresses have been well investigated, glucose deprivation, which can arise from choriocapillary flow voids, has yet to be fully explored. In this study, we examined the involvement of VEGFR2 in glucose deprivation-mediated cell death and the underlying mechanisms. We found that VEGFR2 levels are a determinant for RPE cell death, a critical factor for dry AMD, under glucose deprivation. RNA sequencing analysis showed that upon VEGFR2 knockdown under glucose starvation, endoplasmic reticulum (ER) stress and unfolded protein response (UPR) are reduced. Consistently, VEGFR2 overexpression increased ER stress under the same condition. Although VEGFR2 was less expressed compared to EGFR1 and c-Met in RPE cells, it could elicit a higher level of ER stress induced by glucose starvation. Finally, downregulated VEGFR2 attenuated the oxidative stress and inflammatory factor expression, two downstream targets of ER stress. Our study, for the first time, has demonstrated a novel role of VEGFR2 in RPE cells under glucose deprivation, thus providing valuable insights into the mechanisms of AMD pathogenesis and suggesting that VEGFR2 might be a potential therapeutic target for AMD prevention, which may impede its progression.

2.
Arterioscler Thromb Vasc Biol ; 39(7): 1402-1418, 2019 07.
Article En | MEDLINE | ID: mdl-31242036

Objective- Pathological neovascularization is crucial for progression and morbidity of serious diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. While mechanisms of ongoing pathological neovascularization have been extensively studied, the initiating pathological vascular remodeling (PVR) events, which precede neovascularization remains poorly understood. Here, we identify novel molecular and cellular mechanisms of preneovascular PVR, by using the adult choriocapillaris as a model. Approach and Results- Using hypoxia or forced overexpression of VEGF (vascular endothelial growth factor) in the subretinal space to induce PVR in zebrafish and rats respectively, and by analyzing choriocapillaris membranes adjacent to choroidal neovascular lesions from age-related macular degeneration patients, we show that the choriocapillaris undergo robust induction of vascular intussusception and permeability at preneovascular stages of PVR. This PVR response included endothelial cell proliferation, formation of endothelial luminal processes, extensive vesiculation and thickening of the endothelium, degradation of collagen fibers, and splitting of existing extravascular columns. RNA-sequencing established a role for endothelial tight junction disruption, cytoskeletal remodeling, vesicle- and cilium biogenesis in this process. Mechanistically, using genetic gain- and loss-of-function zebrafish models and analysis of primary human choriocapillaris endothelial cells, we determined that HIF (hypoxia-induced factor)-1α-VEGF-A-VEGFR2 signaling was important for hypoxia-induced PVR. Conclusions- Our findings reveal that PVR involving intussusception and splitting of extravascular columns, endothelial proliferation, vesiculation, fenestration, and thickening is induced before neovascularization, suggesting that identifying and targeting these processes may prevent development of advanced neovascular disease in the future. Visual Overview- An online visual overview is available for this article.


Neovascularization, Pathologic/etiology , Vascular Remodeling/physiology , Adult , Animals , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Macular Degeneration/etiology , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish
3.
Front Immunol ; 9: 1767, 2018.
Article En | MEDLINE | ID: mdl-30105032

Fat is a major tissue component in human breast cancer (BC). Whether breast adipocytes (BAd) affect early stages of BC metastasis is yet unknown. BC progression is dependent on angiogenesis and inflammation, and interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF) are key regulators of these events. Here, we show that BAd increased the dissemination of estrogen receptor positive BC cells (BCC) in vivo in the zebrafish model of metastasis, while dissemination of the more aggressive and metastatic BCC such as estrogen receptor negative was unaffected. While anti-VEGF and anti-IL-8 exhibited equal inhibition of angiogenesis at the primary tumor site, anti-IL-8 reduced BCC dissemination whereas anti-VEGF had minor effects on this early metastatic event. Mechanistically, overexpression of cell-adhesion molecules in BCC and neutrophils via IL-8 increased the dissemination of BCC. Importantly, the extracellular in vivo levels of IL-8 were 40-fold higher than those of VEGF in human BC. Our results suggest that IL-8 is a clinical relevant and promising therapeutic target for human BC.


Adipocytes/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Interleukin-8/metabolism , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Immunological/pharmacology , Biomarkers , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Interleukin-8/antagonists & inhibitors , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neovascularization, Pathologic , Neutrophil Infiltration , Tumor Burden , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Zebrafish
4.
Cancer Immunol Res ; 5(3): 234-247, 2017 03.
Article En | MEDLINE | ID: mdl-28159748

Estradiol (E2) plays a key role in breast cancer progression. Most breast cancer recurrences express the estrogen receptor (ER), but nearly 50% of patients are resistant to antiestrogen therapy. Novel therapeutic targets of ER-positive breast cancers are needed. Protumoral neutrophils expressing the lymphocyte function-associated antigen 1 (LFA-1) integrin may mediate cancer metastasis, and TGFß1 is the major chemoattractant for neutrophils. The role of E2 in neutrophil-ER+ breast cancer cell interactions is unknown. We studied this in vivo using murine breast cancers in immunocompetent mice and human breast cancers in nude mice. Cell dissemination was evaluated in a zebrafish model, and microdialysis of breast cancer patients was performed. In vitro studies were done with mammosphere cultures of breast cancer cells and human neutrophils. We found that E2 increased the number of LFA-1+ neutrophils recruited to the invasive edge of mouse tumors, increased TGFß1 secretion and promoted neutrophil infiltration in mammospheres, and induced overexpression of LFA-1 in neutrophils. In zebrafish, in the presence of E2, neutrophils increased dissemination of ER+ breast cancer cells via LFA-1 and TGFß1, thus causing noninvasive cancer cells to be highly metastatic. Time-lapse imaging in zebrafish revealed close interactions of neutrophils with cancer cells, which drove breast cancer metastasis. We also found that extracellular TGFß1 was overproduced in human breast cancer tissue compared with adjacent normal breast tissue. Thus, E2 can regulate immune/cancer cell interactions in tumor microenvironments. Our results indicate that extracellular TGFß1 is a relevant target in human breast cancer. Cancer Immunol Res; 5(3); 234-47. ©2017 AACR.


Breast Neoplasms/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Estradiol/pharmacology , Neutrophils/drug effects , Neutrophils/physiology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Gene Expression , Humans , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Neoplasm Metastasis , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Burden , Xenograft Model Antitumor Assays , Zebrafish
5.
Sci Data ; 3: 160103, 2016 11 22.
Article En | MEDLINE | ID: mdl-27874850

In angiogenesis with concurrent inflammation, many pathways are activated, some linked to VEGF and others largely VEGF-independent. Pathways involving inflammatory mediators, chemokines, and micro-RNAs may play important roles in maintaining a pro-angiogenic environment or mediating angiogenic regression. Here, we describe a gene expression dataset to facilitate exploration of pro-angiogenic, pro-inflammatory, and remodelling/normalization-associated genes during both an active capillary sprouting phase, and in the restoration of an avascular phenotype. The dataset was generated by microarray analysis of the whole transcriptome in a rat model of suture-induced inflammatory corneal neovascularisation. Regions of active capillary sprout growth or regression in the cornea were harvested and total RNA extracted from four biological replicates per group. High quality RNA was obtained for gene expression analysis using microarrays. Fold change of selected genes was validated by qPCR, and protein expression was evaluated by immunohistochemistry. We provide a gene expression dataset that may be re-used to investigate corneal neovascularisation, and may also have implications in other contexts of inflammation-mediated angiogenesis.


Corneal Neovascularization/genetics , Gene Expression , Animals , Disease Models, Animal , Genome , Microarray Analysis , Neovascularization, Pathologic , Rats
6.
Oncotarget ; 7(49): 81634-81644, 2016 Dec 06.
Article En | MEDLINE | ID: mdl-27835571

BACKGROUND: Radiotherapy is a well-established anti-cancer treatment. Although radiotherapy has been shown to significantly decrease the local relapse in rectal cancer patients, the rate of distant metastasis is still very high. The aim of this study was to evaluate whether AEG-1 is involved in radiation-enhanced migration and invasion in vitro and in a novel in vivo zebrafish model. RESULTS: Migration and invasion were decreased in all the AEG-1 knockdown cell lines. Furthermore, we observed that radiation enhanced migration and invasion, while AEG-1 knockdown abolished this effect. The results from the zebrafish embryo model confirmed the results obtained in vitro. MMP-9 secretion and expression were decreased in AEG-1 knockdown cells. MATERIALS AND METHODS: We evaluated the involvement of AEG-1 in migration and invasion and, radiation-enhanced migration and invasion by Boyden chamber assay in three colon cancer cell lines and respective stable AEG-1 knockdown cell lines. Furthermore, we injected those cells into zebrafish embryos and evaluated the amount of disseminated cells into the tail. CONCLUSION: AEG-1 knockdown inhibits migration and invasion, as well as radiation-enhanced invasion both in vitro and in vivo. We speculate that this is done via the downregulation of the intrinsic or radiation-enhanced MMP-9 expression by AEG-1 in the cancer cells. This study also shows, for the first time, that the zebrafish is a great model to study the early events in radiation-enhanced invasion.


Cell Adhesion Molecules/metabolism , Cell Movement/radiation effects , Colonic Neoplasms/radiotherapy , Gene Knockdown Techniques , Radiation Tolerance , Animals , Cell Adhesion Molecules/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Matrix Metalloproteinase 9/metabolism , Membrane Proteins , Neoplasm Invasiveness , RNA Interference , RNA-Binding Proteins , Signal Transduction/radiation effects , Transfection , Zebrafish/embryology
7.
Biomaterials ; 63: 1-13, 2015 Sep.
Article En | MEDLINE | ID: mdl-26066004

Ultra-small nanoparticles (USNPs) at 1-3 nm are a subset of nanoparticles (NPs) that exhibit intermediate physicochemical properties between molecular dispersions and larger NPs. Despite interest in their utilization in applications such as theranostics, limited data about their toxicity exist. Here the effect of TiO2-USNPs on endothelial cells in vitro, and zebrafish embryos in vivo, was studied and compared to larger TiO2-NPs (30 nm) and to single walled carbon nanotubes (SWCNTs). In vitro exposure showed that TiO2-USNPs were neither cytotoxic, nor had oxidative ability, nevertheless were genotoxic. In vivo experiment in early developing zebrafish embryos in water at high concentrations of TiO2-USNPs caused mortality possibly by acidifying the water and caused malformations in the form of pericardial edema when injected. Myo1C involved in glomerular development of zebrafish embryos was upregulated in embryos exposed to TiO2-USNPs. They also exhibited anti-angiogenic effects both in vitro and in vivo plus decreased nitric oxide concentration. The larger TiO2-NPs were genotoxic but not cytotoxic. SWCNTs were cytotoxic in vitro and had the highest oxidative ability. Neither of these NPs had significant effects in vivo. To our knowledge this is the first study evaluating the effects of TiO2-USNPs on vascular toxicity in vitro and in vivo and this strategy could unravel USNPs potential applications.


Embryo, Nonmammalian/drug effects , Endothelial Cells/drug effects , Nanoparticles/toxicity , Nanotubes, Carbon/toxicity , Neovascularization, Physiologic/drug effects , Titanium/toxicity , Zebrafish/embryology , Animals , Cell Line , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/blood supply , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation, Developmental/drug effects , Humans , Mutagens/chemistry , Mutagens/toxicity , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Nanotubes, Carbon/chemistry , Nanotubes, Carbon/ultrastructure , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Titanium/chemistry
8.
Biochim Biophys Acta ; 1850(8): 1633-41, 2015 Aug.
Article En | MEDLINE | ID: mdl-25450175

BACKGROUND: Cancer is considered to develop due to disruptions in the tissue microenvironment in addition to genetic disruptions in the tumor cells themselves. The two most important microenvironmental disruptions in cancer are arguably tissue hypoxia and disrupted circadian rhythmicity. Endothelial cells, which line the luminal side of all blood vessels transport oxygen or endocrine circadian regulators to the tissue and are therefore of key importance for circadian disruption and hypoxia in tumors. SCOPE OF REVIEW: Here I review recent findings on the role of circadian rhythms and hypoxia in cancer and metastasis, with particular emphasis on how these pathways link tumor metastasis to pathological functions of blood vessels. The involvement of disrupted cell metabolism and redox homeostasis in this context and the use of novel zebrafish models for such studies will be discussed. MAJOR CONCLUSIONS: Circadian rhythms and hypoxia are involved in tumor metastasis on all levels from pathological deregulation of the cell to the tissue and the whole organism. Pathological tumor blood vessels cause hypoxia and disruption in circadian rhythmicity which in turn drives tumor metastasis. Zebrafish models may be used to increase our understanding of the mechanisms behind hypoxia and circadian regulation of metastasis. GENERAL SIGNIFICANCE: Disrupted blood flow in tumors is currently seen as a therapeutic goal in cancer treatment, but may drive invasion and metastasis via pathological hypoxia and circadian clock signaling. Understanding the molecular details behind such regulation is important to optimize treatment for patients with solid tumors in the future. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.


Cell Dedifferentiation/physiology , Circadian Clocks/physiology , Hypoxia/physiopathology , Neoplasms/physiopathology , Animals , Humans , Models, Biological , Neoplasm Metastasis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/physiopathology , Signal Transduction
9.
Biomol Concepts ; 5(3): 245-56, 2014 Jun.
Article En | MEDLINE | ID: mdl-25372756

Daily rhythms of light/darkness, activity/rest and feeding/fasting are important in human physiology and their disruption (for example by frequent changes between day and night shifts) increases the risk of disease. Many of the diseases found to be associated with such disrupted circadian lifestyles, including cancer, cardiovascular diseases, metabolic disorders and neurological diseases, depend on pathological de-regulation of angiogenesis, suggesting that disrupting the circadian clock will impair the physiological regulation of angiogenesis leading to development and progression of these diseases. Today there is little known regarding circadian regulation of pathological angiogenesis but there is some evidence that supports both direct and indirect regulation of angiogenic factors by the cellular circadian clock machinery, as well as by circulating circadian factors, important for coordinating circadian rhythms in the organism. Through highlighting recent advances both in pre-clinical and clinical research on various diseases including cancer, cardiovascular disorders and obesity, we will here present an overview of the available knowledge on the importance of circadian regulation of angiogenesis and discuss how the circadian clock may provide alternative targets for pro- or anti-angiogenic therapy in the future.


Circadian Clocks , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Circadian Rhythm , Gene Expression Regulation , Humans , Mice
10.
Neoplasia ; 16(6): 529-42, 2014 Jun.
Article En | MEDLINE | ID: mdl-25030625

Pigment epithelium-derived factor (PEDF), a member of the serine protease inhibitor superfamily, has potent anti-metastatic effects in cutaneous melanoma through its direct actions on endothelial and melanoma cells. Here we show that PEDF expression positively correlates with microphthalmia-associated transcription factor (MITF) in melanoma cell lines and human samples. High PEDF and MITF expression is characteristic of low aggressive melanomas classified according to molecular and pathological criteria, whereas both factors are decreased in senescent melanocytes and naevi. Importantly, MITF silencing down-regulates PEDF expression in melanoma cell lines and primary melanocytes, suggesting that the correlation in the expression reflects a causal relationship. In agreement, analysis of Chromatin immunoprecipitation coupled to high throughput sequencing (ChIP-seq) data sets revealed three MITF binding regions within the first intron of SERPINF1, and reporter assays demonstrated that the binding of MITF to these regions is sufficient to drive transcription. Finally, we demonstrate that exogenous PEDF expression efficiently halts in vitro migration and invasion, as well as in vivo dissemination of melanoma cells induced by MITF silencing. In summary, these results identify PEDF as a novel transcriptional target of MITF and support a relevant functional role for the MITF-PEDF axis in the biology of melanoma.


Eye Proteins/genetics , Melanoma/genetics , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Nerve Growth Factors/genetics , Serpins/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cellular Senescence/genetics , Disease Progression , Epistasis, Genetic , Eye Proteins/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Metastasis , Nerve Growth Factors/metabolism , Serpins/metabolism
11.
Proc Natl Acad Sci U S A ; 110(50): 20057-62, 2013 Dec 10.
Article En | MEDLINE | ID: mdl-24277839

Embryonic development depends on complex and precisely orchestrated signaling pathways including specific reduction/oxidation cascades. Oxidoreductases of the thioredoxin family are key players conveying redox signals through reversible posttranslational modifications of protein thiols. The importance of this protein family during embryogenesis has recently been exemplified for glutaredoxin 2, a vertebrate-specific glutathione-disulfide oxidoreductase with a critical role for embryonic brain development. Here, we discovered an essential function of glutaredoxin 2 during vascular development. Confocal microscopy and time-lapse studies based on two-photon microscopy revealed that morpholino-based knockdown of glutaredoxin 2 in zebrafish, a model organism to study vertebrate embryogenesis, resulted in a delayed and disordered blood vessel network. We were able to show that formation of a functional vascular system requires glutaredoxin 2-dependent reversible S-glutathionylation of the NAD(+)-dependent protein deacetylase sirtuin 1. Using mass spectrometry, we identified a cysteine residue in the conserved catalytic region of sirtuin 1 as target for glutaredoxin 2-specific deglutathionylation. Thereby, glutaredoxin 2-mediated redox regulation controls enzymatic activity of sirtuin 1, a mechanism we found to be conserved between zebrafish and humans. These results link S-glutathionylation to vertebrate development and successful embryonic angiogenesis.


Cardiovascular System/embryology , Glutaredoxins/metabolism , Glutathione/metabolism , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Sirtuin 1/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Gene Knockdown Techniques , Glutaredoxins/genetics , HeLa Cells , Humans , Mass Spectrometry , Microscopy, Confocal , Oxidation-Reduction , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Time-Lapse Imaging , Zebrafish
12.
Cell Cycle ; 12(3): 405-8, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23324349

Circadian rhythms control multiple physiological and pathological processes, including embryonic development in mammals and development of various human diseases. We have recently, in a developing zebrafish embryonic model, discovered that the circadian oscillation controls developmental angiogenesis. Disruption of crucial circadian regulatory genes, including Bmal1 and Period2, results in marked impairment or enhancement of vascular development in zebrafish. At the molecular level, we show that the circadian regulator Bmal1 directly targets the promoter region of the vegf gene in zebrafish, leading to an elevated expression of VEGF. These findings can reasonably be extended to developmental angiogenesis in mammals and even pathological angiogenesis in humans. Thus, our findings, for the first time, shed new light on mechanisms that underlie circadian clock-regulated angiogenesis.


ARNTL Transcription Factors/genetics , Circadian Rhythm/genetics , Neovascularization, Physiologic/genetics , Period Circadian Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Circadian Clocks , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neovascularization, Physiologic/physiology , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/genetics , Zebrafish/embryology , Zebrafish/genetics
13.
Cell Rep ; 2(2): 231-41, 2012 Aug 30.
Article En | MEDLINE | ID: mdl-22884368

Molecular mechanisms underlying circadian-regulated physiological processes remain largely unknown. Here, we show that disruption of the circadian clock by both constant exposure to light and genetic manipulation of key genes in zebrafish led to impaired developmental angiogenesis. A bmal1-specific morpholino inhibited developmental angiogenesis in zebrafish embryos without causing obvious nonvascular phenotypes. Conversely, a period2 morpholino accelerated angiogenic vessel growth, suggesting that Bmal1 and Period2 display opposing angiogenic effects. Using a promoter-reporter system consisting of various deleted vegf-promoter mutants, we show that Bmal1 directly binds to and activates the vegf promoter via E-boxes. Additionally, we provide evidence that knockdown of Bmal1 leads to impaired Notch-inhibition-induced vascular sprouting. These results shed mechanistic insight on the role of the circadian clock in regulation of developmental angiogenesis, and our findings may be reasonably extended to other types of physiological or pathological angiogenesis.


ARNTL Transcription Factors/metabolism , Neovascularization, Physiologic/physiology , Period Circadian Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , ARNTL Transcription Factors/genetics , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Period Circadian Proteins/genetics , Response Elements/physiology , Vascular Endothelial Growth Factor A/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Nat Med ; 18(1): 100-10, 2011 Dec 04.
Article En | MEDLINE | ID: mdl-22138754

The platelet-derived growth factor (PDGF) signaling system contributes to tumor angiogenesis and vascular remodeling. Here we show in mouse tumor models that PDGF-BB induces erythropoietin (EPO) mRNA and protein expression by targeting stromal and perivascular cells that express PDGF receptor-ß (PDGFR-ß). Tumor-derived PDGF-BB promoted tumor growth, angiogenesis and extramedullary hematopoiesis at least in part through modulation of EPO expression. Moreover, adenoviral delivery of PDGF-BB to tumor-free mice increased both EPO production and erythropoiesis, as well as protecting from irradiation-induced anemia. At the molecular level, we show that the PDGF-BB-PDGFR-bß signaling system activates the EPO promoter, acting in part through transcriptional regulation by the transcription factor Atf3, possibly through its association with two additional transcription factors, c-Jun and Sp1. Our findings suggest that PDGF-BB-induced EPO promotes tumor growth through two mechanisms: first, paracrine stimulation of tumor angiogenesis by direct induction of endothelial cell proliferation, migration, sprouting and tube formation, and second, endocrine stimulation of extramedullary hematopoiesis leading to increased oxygen perfusion and protection against tumor-associated anemia.


Erythropoietin/metabolism , Hematopoiesis , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Anemia/genetics , Animals , Becaplermin , Carcinoma, Lewis Lung/blood supply , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Erythropoietin/genetics , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred C57BL , Mice, SCID , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-sis/genetics , Signal Transduction , Stromal Cells/metabolism
15.
Birth Defects Res C Embryo Today ; 93(2): 182-93, 2011 Jun.
Article En | MEDLINE | ID: mdl-21671357

Most in vivo preclinical disease models are based on mouse and other mammalian systems. However, these rodent-based model systems have considerable limitations to recapitulate clinical situations in human patients. Zebrafish have been widely used to study embryonic development, behavior, tissue regeneration, and genetic defects. Additionally, zebrafish also provides an opportunity to screen chemical compounds that target a specific cell population for drug development. Owing to the availability of various genetically manipulated strains of zebrafish, immune privilege during early embryonic development, transparency of the embryos, and easy and precise setup of hypoxia equipment, we have developed several disease models in both embryonic and adult zebrafish, focusing on studying the role of angiogenesis in pathological settings. These zebrafish disease models are complementary to the existing mouse models, allowing us to study clinically relevant processes in cancer and nonmalignant diseases, which otherwise would be difficult to study in mice. For example, dissemination and invasion of single human or mouse tumor cells from the primary site in association with tumor angiogenesis can be studied under normoxia or hypoxia in zebrafish embryos. Hypoxia-induced retinopathy in the adult zebrafish recapitulates the clinical situation of retinopathy development in diabetic patients or age-related macular degeneration. These zebrafish disease models offer exciting opportunities to understand the mechanisms of disease development, progression, and development of more effective drugs for therapeutic intervention.


Cardiovascular System/embryology , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Lymphatic System/embryology , Macular Degeneration/physiopathology , Neoplasms/physiopathology , Neovascularization, Pathologic/physiopathology , Zebrafish , Animals , Animals, Genetically Modified , Cardiovascular System/anatomy & histology , Cell Hypoxia/physiology , Humans , Lymphatic System/anatomy & histology , Lymphatic System/physiology , Neovascularization, Pathologic/etiology , Regeneration/physiology , Signal Transduction/physiology , Species Specificity
16.
Cell Cycle ; 9(5): 913-7, 2010 Mar 01.
Article En | MEDLINE | ID: mdl-20160500

Clinically detectable metastases represent an ultimate consequence of the metastatic cascade that consists of distinct processes including tumor cell invasion, dissemination, metastatic niche formation, and re-growth into a detectable metastatic mass. Although angiogenesis is known to promote tumor growth, its role in facilitating early events of the metastatic cascade remains poorly understood. We have recently developed a zebrafish tumor model that enables us to study involvement of pathological angiogenesis in tumor invasion, dissemination and metastasis. This non-invasive in vivo model allows detection of single malignant cell dissemination under both normoxia and hypoxia. Further, hypoxia-induced VEGF significantly facilitates tumor cell invasion and dissemination. These findings demonstrate that VEGF-induced pathological angiogenesis is essential for tumor dissemination and further corroborates potentially beneficial effects of clinically ongoing anti-VEGF drugs for the treatment of various malignancies.


Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Animals , Hypoxia , Models, Animal , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
17.
PLoS One ; 3(7): e2748, 2008 Jul 23.
Article En | MEDLINE | ID: mdl-18648503

Mechanistic understanding and defining novel therapeutic targets of diabetic retinopathy and age-related macular degeneration (AMD) have been hampered by a lack of appropriate adult animal models. Here we describe a simple and highly reproducible adult fli-EGFP transgenic zebrafish model to study retinal angiogenesis. The retinal vasculature in the adult zebrafish is highly organized and hypoxia-induced neovascularization occurs in a predictable area of capillary plexuses. New retinal vessels and vascular sprouts can be accurately measured and quantified. Orally active anti-VEGF agents including sunitinib and ZM323881 effectively block hypoxia-induced retinal neovascularization. Intriguingly, blockage of the Notch signaling pathway by the inhibitor DAPT under hypoxia, results in a high density of arterial sprouting in all optical arteries. The Notch suppression-induced arterial sprouting is dependent on tissue hypoxia. However, in the presence of DAPT substantial endothelial tip cell formation was detected only in optic capillary plexuses under normoxia. These findings suggest that hypoxia shifts the vascular targets of Notch inhibitors. Our findings for the first time show a clinically relevant retinal angiogenesis model in adult zebrafish, which might serve as a platform for studying mechanisms of retinal angiogenesis, for defining novel therapeutic targets, and for screening of novel antiangiogenic drugs.


Hypoxia , Neovascularization, Pathologic , Retinal Diseases/diagnosis , Retinal Diseases/pathology , Retinal Neovascularization/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Humans , Indoles/pharmacology , Microscopy, Confocal , Pyrroles/pharmacology , Receptors, Notch/metabolism , Retinal Neovascularization/metabolism , Signal Transduction , Sunitinib , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
...