Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Transl Res ; 255: 97-108, 2023 05.
Article in English | MEDLINE | ID: mdl-36481562

ABSTRACT

Accurately modeling tumor biology and testing novel therapies on patient-derived cells is critically important to developing therapeutic regimens personalized to a patient's specific disease. The vascularized microtumor (VMT), or "tumor-on-a-chip," is a physiologic preclinical cancer model that incorporates key features of the native human tumor microenvironment within a transparent microfluidic platform, allowing rapid drug screening in vitro. Herein we optimize methods for generating patient-derived VMT (pVMT) using fresh colorectal cancer (CRC) biopsies and surgical resections to test drug sensitivities at the individual patient level. In response to standard chemotherapy and TGF-ßR1 inhibition, we observe heterogeneous responses between pVMT derived from 6 patient biopsies, with the pVMT recapitulating tumor growth, histological features, metabolic heterogeneity, and drug responses of actual CRC tumors. Our results suggest that a translational infrastructure providing rapid information from patient-derived tumor cells in the pVMT, as established in this study, will support efforts to improve patient outcomes.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Microfluidics , Tumor Microenvironment
2.
PLoS One ; 15(12): e0242543, 2020.
Article in English | MEDLINE | ID: mdl-33326419

ABSTRACT

Clinical studies using a range of omega-3 supplements have yielded conflicting results on their efficacy to control inflammation. Omega-3 fatty acids are substrate for the formation of potent immune-protective mediators, termed as specialized pro-resolving mediators (SPM). Herein, we investigated whether observed differences in the potencies of distinct omega-3 supplements were linked with their ability to upregulate SPM formation. Using lipid mediator profiling we found that four commercially available supplements conferred a unique SPM signature profile to human macrophages, with the overall increases in SPM concentrations being different between the four supplements. These increases in SPM concentrations were linked with an upregulation of macrophage phagocytosis and a decreased uptake of oxidized low-density lipoproteins. Pharmacological inhibition of two key SPM biosynthetic enzymes 5-Lipoxygenase or 15-Lipoxygenase reversed the macrophage-directed actions of each of the omega-3 supplements. Furthermore, administration of the two supplements that most potently upregulated macrophage SPM formation and reprogrammed their responses in vitro, to APOE-/- mice fed a western diet, increased plasma SPM concentrations and reduced vascular inflammation. Together these findings support the utility of SPM as potential prognostic markers in determining the utility of a given supplement to regulate macrophage responses and inflammation.


Subject(s)
Atherosclerosis/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/administration & dosage , Leukotrienes/biosynthesis , Lipoxins/biosynthesis , Macrophages/drug effects , Prostaglandins/biosynthesis , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/immunology , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/immunology , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Diet, Western/adverse effects , Fatty Acids, Omega-3/metabolism , Female , Gene Expression , Humans , Leukotrienes/immunology , Lipoproteins, LDL/antagonists & inhibitors , Lipoproteins, LDL/pharmacology , Lipoxins/immunology , Lipoxygenase Inhibitors/pharmacology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Knockout, ApoE , Phagocytosis/drug effects , Primary Cell Culture , Principal Component Analysis , Prostaglandins/immunology
3.
J Clin Invest ; 130(1): 359-373, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31793912

ABSTRACT

N-3 docosapentaenoic acid-derived resolvin D5 (RvD5n-3 DPA) is diurnally regulated in peripheral blood and exerts tissue-protective actions during inflammatory arthritis. Here, using an orphan GPCR screening approach coupled with functional readouts, we investigated the receptor(s) involved in mediating the leukocyte-directed actions of RvD5n-3 DPA and identified GPR101 as the top candidate. RvD5n-3 DPA bound to GPR101 with high selectivity and stereospecificity, as demonstrated by a calculated KD of approximately 6.9 nM. In macrophages, GPR101 knockdown limited the ability of RvD5n-3 DPA to upregulate cyclic adenosine monophosphate, phagocytosis of bacteria, and efferocytosis. Inhibition of this receptor in mouse and human leukocytes abrogated the pro-resolving actions of RvD5n-3 DPA, including the regulation of bacterial phagocytosis in neutrophils. Knockdown of the receptor in vivo reversed the protective actions of RvD5n-3 DPA in limiting joint and gut inflammation during inflammatory arthritis. Administration of RvD5n-3 DPA during E. coli-initiated inflammation regulated neutrophil trafficking to the site of inflammation, increased bacterial phagocytosis by neutrophils and macrophages, and accelerated the resolution of infectious inflammation. These in vivo protective actions of RvD5n-3 DPA were limited when Gpr101 was knocked down. Together, our findings demonstrate a fundamental role for GPR101 in mediating the leukocyte-directed actions of RvD5n-3 DPA.


Subject(s)
Arthritis/drug therapy , Docosahexaenoic Acids/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli/immunology , Macrophages/immunology , Neutrophils/immunology , Receptors, G-Protein-Coupled/agonists , Animals , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , CHO Cells , Cricetulus , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Gene Knockdown Techniques , Humans , Macrophages/pathology , Male , Mice , Neutrophils/pathology , Phagocytosis/drug effects , Phagocytosis/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology
4.
IEEE/ACM Trans Comput Biol Bioinform ; 16(3): 1029-1035, 2019.
Article in English | MEDLINE | ID: mdl-29993583

ABSTRACT

Likely drug candidates which are identified in traditional pre-clinical drug screens often fail in patient trials, increasing the societal burden of drug discovery. A major contributing factor to this phenomenon is the failure of traditional in vitro models of drug response to accurately mimic many of the more complex properties of human biology. We have recently introduced a new microphysiological system for growing vascularized, perfused microtissues that more accurately models human physiology and is suitable for large drug screens. In this work, we develop a machine learning model that can quickly and accurately flag compounds which effectively disrupt vascular networks from images taken before and after drug application in vitro. The system is based on a convolutional neural network and achieves near perfect accuracy while committing potentially no expensive false negatives.


Subject(s)
Antineoplastic Agents/pharmacology , Deep Learning , Drug Discovery/methods , Image Processing, Computer-Assisted , Neoplasms/drug therapy , Neovascularization, Pathologic/diagnostic imaging , Cell Culture Techniques , Extracellular Matrix/metabolism , Humans , Microscopy , Neoplasms/diagnostic imaging , Neural Networks, Computer , Pattern Recognition, Automated
5.
Exp Biol Med (Maywood) ; 242(17): 1669-1678, 2017 11.
Article in English | MEDLINE | ID: mdl-28195514

ABSTRACT

The blood-brain barrier is a dynamic and highly organized structure that strictly regulates the molecules allowed to cross the brain vasculature into the central nervous system. The blood-brain barrier pathology has been associated with a number of central nervous system diseases, including vascular malformations, stroke/vascular dementia, Alzheimer's disease, multiple sclerosis, and various neurological tumors including glioblastoma multiforme. There is a compelling need for representative models of this critical interface. Current research relies heavily on animal models (mostly mice) or on two-dimensional (2D) in vitro models, neither of which fully capture the complexities of the human blood-brain barrier. Physiological differences between humans and mice make translation to the clinic problematic, while monolayer cultures cannot capture the inherently three-dimensional (3D) nature of the blood-brain barrier, which includes close association of the abluminal side of the endothelium with astrocyte foot-processes and pericytes. Here we discuss the central nervous system diseases associated with blood-brain barrier pathology, recent advances in the development of novel 3D blood-brain barrier -on-a-chip systems that better mimic the physiological complexity and structure of human blood-brain barrier, and provide an outlook on how these blood-brain barrier-on-a-chip systems can be used for central nervous system disease modeling. Impact statement The field of microphysiological systems is rapidly evolving as new technologies are introduced and our understanding of organ physiology develops. In this review, we focus on Blood-Brain Barrier (BBB) models, with a particular emphasis on how they relate to neurological disorders such as Alzheimer's disease, multiple sclerosis, stroke, cancer, and vascular malformations. We emphasize the importance of capturing the three-dimensional nature of the brain and the unique architecture of the BBB - something that until recently had not been well modeled by in vitro systems. Our hope is that this review will provide a launch pad for new ideas and methodologies that can provide us with truly physiological BBB models capable of yielding new insights into the function of this critical interface.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain/blood supply , Endothelium, Vascular/metabolism , Microchip Analytical Procedures/methods , Microtechnology/methods , Tissue Engineering/methods , Alzheimer Disease/pathology , Biological Transport/physiology , Glioblastoma/pathology , Humans , Lab-On-A-Chip Devices , Models, Biological , Multiple Sclerosis/pathology , Stroke/pathology
6.
Lab Chip ; 17(3): 511-520, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28092382

ABSTRACT

There is a growing awareness that complex 3-dimensional (3D) organs are not well represented by monolayers of a single cell type - the standard format for many drug screens. To address this deficiency, and with the goal of improving screens so that drugs with good efficacy and low toxicity can be identified, microphysiological systems (MPS) are being developed that better capture the complexity of in vivo physiology. We have previously described an organ-on-a-chip platform that incorporates perfused microvessels, such that survival of the surrounding tissue is entirely dependent on delivery of nutrients through the vessels. Here we describe an arrayed version of the platform that incorporates multiple vascularized micro-organs (VMOs) on a 96-well plate. Each VMO is independently-addressable and flow through the micro-organ is driven by hydrostatic pressure. The platform is easy to use, requires no external pumps or valves, and is highly reproducible. As a proof-of-concept we have created arrayed vascularized micro tumors (VMTs) and used these in a blinded screen to assay a small library of compounds, including FDA-approved anti-cancer drugs, and successfully identified both anti-angiogenic and anti-tumor drugs. This 3D platform is suitable for efficacy/toxicity screening against multiple tissues in a more physiological environment than previously possible.


Subject(s)
Cell Culture Techniques/instrumentation , Drug Evaluation, Preclinical/instrumentation , Microfluidic Analytical Techniques/instrumentation , Tissue Array Analysis/instrumentation , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical/methods , Equipment Design , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/metabolism , Tissue Array Analysis/methods
7.
Biochem Pharmacol ; 129: 67-72, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28131844

ABSTRACT

The Mas receptor is involved in the angiotensin (Ang)-(1-7) vasodilatory actions by increasing nitric oxide production (NO). We have previously demonstrated an increased production of Ang-(1-7) in human umbilical vein endothelial cells (HUVEC) exposed to estradiol (E2), suggesting a potential cross-talk between E2 and the Ang-(1-7)/Mas receptor axis. Here, we explored whether the vasoactive response and NO-related signalling exerted by E2 are influenced by Mas. HUVEC were exposed to 10nM E2 for 24h in the presence or absence of the selective Mas receptor antagonist A779, and the estrogen receptor (ER) antagonist ICI182780 (ICI). E2 increased Akt and endothelial nitric oxide synthase (eNOS) mRNA and protein expression, measured by RT-PCR and Western blot, respectively. Furthermore, E2 increased Akt activity (determined by the levels of phospho-Ser473) and eNOS activity (by the enhanced phosphorylation of Ser1177, the activated form), resulting in increased NO production, which was measured by the fluorescence probe DAF-2-FM. These signalling events were dependent on ER and Mas receptor activation, since they were abolished in the presence of ICI or A779. In ex-vivo functional experiments performed with a small-vessel myograph in isolated mesenteric vessels from wild-type mice pre-contracted with noradrenaline, the relaxant response to physiological concentrations of E2 was blocked by ICI and A779, to the same extent to that obtained in the vessels isolated from Mas-deficient. In conclusion, E2 induces NO production and vasodilation through mechanisms that require Mas receptor activation.


Subject(s)
Nitric Oxide/physiology , Proto-Oncogene Proteins/physiology , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , Vasodilation/physiology , Animals , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Mas
8.
Biomaterials ; 116: 118-129, 2017 02.
Article in English | MEDLINE | ID: mdl-27914984

ABSTRACT

Extracellular matrix (ECM) is an essential and dynamic component of all tissues and directly affects cellular behavior by providing both mechanical and biochemical signaling cues. Changes in ECM can alter tissue homeostasis, potentially leading to promotion of cellular transformation and the generation of tumors. Therefore, understanding ECM compositional changes during cancer progression is vital to the development of targeted treatments. Previous efforts to reproduce the native 3D cellular microenvironment have utilized protein gels and scaffolds that incompletely recapitulate the complexity of native tissues. Here, we address this problem by extracting and comparing ECM from normal human colon and colon tumor that had metastasized to liver. We found differences in protein composition and stiffness, and observed significant differences in vascular network formation and tumor growth in each of the reconstituted matrices, both in vitro and in vivo. We studied free/bound ratios of NADH in the tumor and endothelial cells using Fluorescence Lifetime Imaging Microscopy as a surrogate for the metabolic state of the cells. We observed that cells seeded in tumor ECM had higher relative levels of free NADH, consistent with a higher glycolytic rate, than those seeded in normal ECM. These results demonstrate that the ECM plays an important role in the growth of cancer cells and their associated vasculature.


Subject(s)
Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Tumor Microenvironment , Cell Proliferation , Colonic Neoplasms/blood supply , Humans , Tumor Cells, Cultured
9.
Sci Rep ; 6: 31589, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27549930

ABSTRACT

There is a growing interest in developing microphysiological systems that can be used to model both normal and pathological human organs in vitro. This "organs-on-chips" approach aims to capture key structural and physiological characteristics of the target tissue. Here we describe in vitro vascularized microtumors (VMTs). This "tumor-on-a-chip" platform incorporates human tumor and stromal cells that grow in a 3D extracellular matrix and that depend for survival on nutrient delivery through living, perfused microvessels. Both colorectal and breast cancer cells grow vigorously in the platform and respond to standard-of-care therapies, showing reduced growth and/or regression. Vascular-targeting agents with different mechanisms of action can also be distinguished, and we find that drugs targeting only VEGFRs (Apatinib and Vandetanib) are not effective, whereas drugs that target VEGFRs, PDGFR and Tie2 (Linifanib and Cabozantinib) do regress the vasculature. Tumors in the VMT show strong metabolic heterogeneity when imaged using NADH Fluorescent Lifetime Imaging Microscopy and, compared to their surrounding stroma, many show a higher free/bound NADH ratio consistent with their known preference for aerobic glycolysis. The VMT platform provides a unique model for studying vascularized solid tumors in vitro.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms , Colorectal Neoplasms , Microfluidic Analytical Techniques , Models, Biological , Neovascularization, Pathologic , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , MCF-7 Cells , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
10.
Tissue Eng Part A ; 22(15-16): 1016-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27392582

ABSTRACT

Pluripotent stem cell-derived cardiomyocytes (CMs) have great potential in the development of new therapies for cardiovascular disease. In particular, human induced pluripotent stem cells (iPSCs) may prove especially advantageous due to their pluripotency, their self-renewal potential, and their ability to create patient-specific cell lines. Unfortunately, pluripotent stem cell-derived CMs are immature, with characteristics more closely resembling fetal CMs than adult CMs, and this immaturity has limited their use in drug screening and cell-based therapies. Extracellular matrix (ECM) influences cellular behavior and maturation, as does the geometry of the environment-two-dimensional (2D) versus three-dimensional (3D). We therefore tested the hypothesis that native cardiac ECM and 3D cultures might enhance the maturation of iPSC-derived CMs in vitro. We demonstrate that maturation of iPSC-derived CMs was enhanced when cells were seeded into a 3D cardiac ECM scaffold, compared with 2D culture. 3D cardiac ECM promoted increased expression of calcium-handling genes, Junctin, CaV1.2, NCX1, HCN4, SERCA2a, Triadin, and CASQ2. Consistent with this, we find that iPSC-derived CMs in 3D adult cardiac ECM show increased calcium signaling (amplitude) and kinetics (maximum upstroke and downstroke) compared with cells in 2D. Cells in 3D culture were also more responsive to caffeine, likely reflecting an increased availability of calcium in the sarcoplasmic reticulum. Taken together, these studies provide novel strategies for maturing iPSC-derived CMs that may have applications in drug screening and transplantation therapies to treat heart disease.


Subject(s)
Antigens, Differentiation/biosynthesis , Extracellular Matrix/chemistry , Induced Pluripotent Stem Cells/metabolism , Myocardium/chemistry , Myocytes, Cardiac/metabolism , Tissue Scaffolds/chemistry , Animals , Cattle , Coculture Techniques
11.
Lab Chip ; 16(2): 282-90, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26616908

ABSTRACT

This paper reports a method for generating an intact and perfusable microvascular network that connects to microfluidic channels without appreciable leakage. This platform incorporates different stages of vascular development including vasculogenesis, endothelial cell (EC) lining, sprouting angiogenesis, and anastomosis in sequential order. After formation of a capillary network inside the tissue chamber via vasculogenesis, the adjacent microfluidic channels are lined with a monolayer of ECs, which then serve as the high-pressure input ("artery") and low pressure output ("vein") conduits. To promote a tight interconnection between the artery/vein and the capillary network, sprouting angiogenesis is induced, which promotes anastomosis of the vasculature inside the tissue chamber with the EC lining along the microfluidic channels. Flow of fluorescent microparticles confirms the perfusability of the lumenized microvascular network, and minimal leakage of 70 kDa FITC-dextran confirms physiologic tightness of the EC junctions and completeness of the interconnections between artery/vein and the capillary network. This versatile device design and its robust construction methodology establish a physiological transport model of interconnected perfused vessels from artery to vascularized tissue to vein. The system has utility in a wide range of organ-on-a-chip applications as it enables the physiological vascular interconnection of multiple on-chip tissue constructs that can serve as disease models for drug screening.


Subject(s)
Endothelial Cells/cytology , Microfluidic Analytical Techniques , Microvessels , Neovascularization, Physiologic , Tissue Engineering , Anastomosis, Surgical , Cells, Cultured , Humans , Microfluidic Analytical Techniques/instrumentation
12.
PLoS One ; 10(12): e0144572, 2015.
Article in English | MEDLINE | ID: mdl-26695765

ABSTRACT

Supervised machine learning can be used to predict which drugs human cardiomyocytes have been exposed to. Using electrophysiological data collected from human cardiomyocytes with known exposure to different drugs, a supervised machine learning algorithm can be trained to recognize and classify cells that have been exposed to an unknown drug. Furthermore, the learning algorithm provides information on the relative contribution of each data parameter to the overall classification. Probabilities and confidence in the accuracy of each classification may also be determined by the algorithm. In this study, the electrophysiological effects of ß-adrenergic drugs, propranolol and isoproterenol, on cardiomyocytes derived from human induced pluripotent stem cells (hiPS-CM) were assessed. The electrophysiological data were collected using high temporal resolution 2-photon microscopy of voltage sensitive dyes as a reporter of membrane voltage. The results demonstrate the ability of our algorithm to accurately assess, classify, and predict hiPS-CM membrane depolarization following exposure to chronotropic drugs.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Heart Rate/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Algorithms , Cell Differentiation/drug effects , Cell Line , Electrophysiologic Techniques, Cardiac , Humans , Induced Pluripotent Stem Cells/cytology , Isoproterenol/pharmacology , Propranolol/pharmacology , Supervised Machine Learning
13.
Exp Biol Med (Maywood) ; 239(9): 1240-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24740872

ABSTRACT

Cancer is one of the leading causes of morbidity and mortality around the world. Despite some success, traditional anticancer drugs developed to reduce tumor growth face important limitations primarily due to undesirable bone marrow and cardiovascular toxicity. Many drugs fail in clinical development after showing promise in preclinical trials, suggesting that the available in vitro and animal models are poor predictors of drug efficacy and toxicity in humans. Thus, novel models that more accurately mimic the biology of human organs are necessary for high-throughput drug screening. Three-dimensional (3D) microphysiological systems can utilize induced pluripotent stem cell technology, tissue engineering, and microfabrication techniques to develop tissue models of human tumors, cardiac muscle, and bone marrow on the order of 1 mm(3) in size. A functional network of human capillaries and microvessels to overcome diffusion limitations in nutrient delivery and waste removal can also nourish the 3D microphysiological tissues. Importantly, the 3D microphysiological tissues are grown on optically clear platforms that offer non-invasive and non-destructive image acquisition with subcellular resolution in real time. Such systems offer a new paradigm for high-throughput drug screening and will significantly improve the efficiency of identifying new drugs for cancer treatment that minimize cardiac and bone marrow toxicity.


Subject(s)
Cell Culture Techniques , Drug Screening Assays, Antitumor , Neoplasms/drug therapy , Neoplasms/metabolism , Pluripotent Stem Cells/metabolism , Tissue Engineering , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Humans , Neoplasms/pathology , Pluripotent Stem Cells/pathology , Tissue Engineering/instrumentation , Tissue Engineering/methods
14.
Mol Cell Endocrinol ; 365(1): 11-6, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-22982060

ABSTRACT

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide (NO) synthase. ADMA accumulation, mainly due to a decreased dimethylarginine dimethylaminohydrolase (DDAH) activity, has been related to the development of cardiovascular diseases. We investigate whether estradiol prevents the changes induced by oxidized low density lipoprotein (oxLDL) on the DDAH/ADMA/NO pathway in human umbilical artery endothelial cells (HUAEC). HUAEC were exposed to estradiol, native LDL (nLDL), oxLDL and their combinations for 24 h. In some experiments, cells were also exposed to the unspecific estrogen receptor (ER) antagonist ICI 182780, the specific ERα antagonist MPP or specific agonists for ERα, ERß and GPER. ADMA concentration was measured by HPLC and concentration of NO by amperometry. Protein expression and DDAH activity were measured by immunoblotting and an enzymatic method, respectively. oxLDL, but not nLDL, increased ADMA concentration with a concomitant decrease on DDAH activity. oxLDL reduced eNOS protein and NO production. Estradiol alone had no effects on DDAH/ADMA/NO pathway, but increased the attenuated endothelial NO production induced by oxLDL by reduction in ADMA and preventing loss of eNOS protein levels. ICI 182780 and MPP completely abolished these effects of estradiol on oxLDL-exposed cells. ERα agonist, but not ERß and GPER agonists, mirrored estradiol effects on NO production. In conclusion, estradiol restores (1) DDAH activity, and therefore ADMA levels, and (2) NO production impaired by oxLDL in HUAEC acting through ERα.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Endothelium, Vascular/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Lipoproteins, LDL/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/agonists , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Arginine/adverse effects , Arginine/analogs & derivatives , Arginine/antagonists & inhibitors , Arginine/metabolism , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Estradiol/agonists , Estradiol/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Estrogens, Non-Steroidal/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Lipoproteins, LDL/adverse effects , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/chemistry , Protein Stability , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Umbilical Arteries/cytology
15.
Mol Cell Endocrinol ; 335(2): 96-103, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-20615453

ABSTRACT

Migration and proliferation of endothelial cells are involved in re-endothelialization and angiogenesis, two important cardiovascular processes that are increased in response to estrogens. RhoA, a small GTPase which controls multiple cellular processes, is involved in the control of cell migration and proliferation. Our aim was to study the role of RhoA on estradiol-induced migration and proliferation and its dependence on estrogen receptors activity. Human umbilical vein endothelial cells were stimulated with estradiol, in the presence or absence of ICI 182780 (estrogen receptors antagonist) and Y-27632 (Rho kinase inhibitor). Estradiol increased Rho GEF-1 gene expression and RhoA (gene and protein expression and activity) in an estrogen receptor-dependent manner. Cell migration, stress fiber formation and cell proliferation were increased in response to estradiol and were also dependent on the estrogen receptors and RhoA activation. Estradiol decreased p27 levels, and significantly raised the expression of cyclins and CDK. These effects were counteracted by the use of either ICI 182780 or Y-27632. In conclusion, estradiol enhances the RhoA/ROCK pathway and increases cell cycle-related protein expression by acting through estrogen receptors. This results in an enhanced migration and proliferation of endothelial cells.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/metabolism , Estradiol/pharmacology , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Assays , Female , Humans , Male , Transcriptional Activation/drug effects , Umbilical Cord/cytology , Up-Regulation/drug effects , rhoA GTP-Binding Protein/genetics
16.
J Mol Endocrinol ; 44(4): 237-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110403

ABSTRACT

Estradiol (E(2)) acts on the endothelium to promote vasodilatation through the release of several compounds, including prostanoids, which are products of arachidonic acid metabolism. Among these, prostacyclin (PGI2) and thromboxane A2 (TXA2) exert opposite effects on vascular tone. The role of different estrogen receptors (ERs) in the PGI2/TXA2 balance, however, has not been fully elucidated. Our study sought to uncover whether E(2) enhances basal production of PGI2 or TXA2 in cultured human umbilical vein endothelial cells (HUVECs), to analyze the enzymatic mechanisms involved, and to evaluate the different roles of both types of ERs (ERalpha and ERbeta). HUVECs were exposed to E(2), selective ERalpha (1,3,5-tris(4-hydroxyphenyl)-4-propyl-1h-pyrazole, PPT) or ERbeta (diarylpropionitrile, DPN) agonists and antagonists (unspecific: ICI 182 780; specific for ERalpha: methyl-piperidino-pyrazole, MPP). PGI2 and TXA2 production was measured by ELISA. Expression of phospholipases, cyclooxygenases (COX-1 and COX-2), PGI2 synthase (PGIS), and thromboxane synthase (TXAS) was analyzed by western blot and quantitative RT-PCR. E(2) (1-100 nM) dose dependently increased PGI2 production (up to 50%), without affecting TXA2 production. COX-1 and PGIS protein and gene expressions were increased, whereas COX-2, phospholipases, and TXAS expression remained unaltered. All these effects were mediated through ERalpha, since they were produced not only in the presence of E(2), but also in that of PPT, while they were abolished in the presence of MPP. In conclusion, E(2), acting through ERalpha, up-regulates COX-1 and PGIS expression, thus directing prostanoid balance toward increased PGI2 production.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Epoprostenol/biosynthesis , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Epoprostenol/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Male , Thromboxane A2/metabolism
17.
PLoS One ; 4(12): e8242, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20011585

ABSTRACT

Vascular effects of estradiol are being investigated because there are controversies among clinical and experimental studies. DNA microarrays were used to investigate global gene expression patterns in cultured human umbilical vein endothelial cells (HUVEC) exposed to 1 nmol/L estradiol for 24 hours. When compared to control, 187 genes were identified as differentially expressed with 1.9-fold change threshold. Supervised principal component analysis and hierarchical cluster analysis revealed the differences between control and estradiol-treated samples. Physiological concentrations of estradiol are sufficient to elicit significant changes in HUVEC gene expression. Notch signaling, actin cytoskeleton signaling, pentose phosphate pathway, axonal guidance signaling and integrin signaling were the top-five canonical pathways significantly regulated by estrogen. A total of 26 regulatory networks were identified as estrogen responsive. Microarray data were confirmed by quantitative RT-PCR in cardiovascular meaning genes; cyclooxygenase (COX)1, dimethylarginine dimethylaminohydrolase (DDAH)2, phospholipase A2 group IV (PLA2G4) B, and 7-dehydrocholesterol reductase were up-regulated by estradiol in a dose-dependent and estrogen receptor-dependent way, whereas COX2, DDAH1 and PLA2G4A remained unaltered. Moreover, estradiol-induced COX1 gene expression resulted in increased COX1 protein content and enhanced prostacyclin production. DDAH2 protein content was also increased, which in turn decreased asymmetric dimethylarginine concentration and increased NO release. All stimulated effects of estradiol on gene and protein expression were estrogen receptor-dependent, since were abolished in the presence of the estrogen receptor antagonist ICI 182780. This study identifies new vascular mechanisms of action by which estradiol may contribute to a wide range of biological processes.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Estradiol/pharmacology , Metabolic Networks and Pathways/drug effects , Vasodilation/drug effects , Amidohydrolases/genetics , Amidohydrolases/metabolism , Cells, Cultured , Cluster Analysis , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Metabolic Networks and Pathways/genetics , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Umbilical Veins/cytology
18.
Gynecol Endocrinol ; 24(6): 354-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18584416

ABSTRACT

The CD40/CD40L system is considered a crucial modulator of the inflammatory process underlying the progression and complication of atheroma plaques. The soluble fraction of CD40L (sCD40L) is a reliable indicator of the CD40/CD40L system. Our purpose was to investigate whether a therapeutic dose of estradiol, by either the oral or the transdermal route, was associated with changes in circulating levels of sCD40L. Forty-seven women completed a 4-week course of treatment with either oral estradiol valerate (2 mg/day, 20 women) or transdermal estradiol (50 microg/day, 27 women). Serum levels of sCD40L were measured by conventional enzyme-linked immunosorbent assay. Oral, but not transdermal estradiol, modified the lipid profile. Levels of sCD40L, however, remained unchanged compared with baseline. This neutral effect of oral or transdermal estradiol on sCD40L levels further advances our knowledge on the effects of estrogens on mechanisms involved in the progression and complication of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , CD40 Antigens/blood , CD40 Ligand/blood , Estradiol/administration & dosage , Administration, Cutaneous , Administration, Oral , Atherosclerosis/blood , CD40 Antigens/immunology , CD40 Ligand/immunology , Cholesterol/blood , Enzyme-Linked Immunosorbent Assay , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Humans , Postmenopause/blood , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...