Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Am J Physiol Cell Physiol ; 319(1): C183-C193, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32432925

ABSTRACT

The vasa vasorum (VV), the microvascular network around large vessels, has been recognized as an important contributor to the pathological vascular remodeling in cardiovascular diseases. In bovine and rat models of hypoxic pulmonary hypertension (PH), we have previously shown that chronic hypoxia profoundly increased pulmonary artery (PA) VV permeability, associated with infiltration of inflammatory and progenitor cells in the arterial wall, perivascular inflammation, and structural vascular remodeling. Extracellular adenosine was shown to exhibit a barrier-protective effect on VV endothelial cells (VVEC) via cAMP-independent mechanisms, which involved adenosine A1 receptor-mediated activation of Gi-phosphoinositide 3-kinase-Akt pathway and actin cytoskeleton remodeling. Using VVEC isolated from the adventitia of calf PA, in this study we investigated in more detail the mechanisms linking Gi activation to downstream barrier protection pathways. Using a small-interference RNA (siRNA) technique and transendothelial electrical resistance assay, we found that the adaptor protein, engulfment and cell motility 1 (ELMO1), the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2, and atypical Gi- and Rac1-mediated protein kinase A activation are implicated in VVEC barrier enhancement. In contrast, the actin-interacting GTP-binding protein, girdin, and the p21-activated kinase 1 downstream target, LIM kinase, are not involved in this response. In addition, adenosine-dependent cytoskeletal rearrangement involves activation of cofilin and inactivation of ezrin-radixin-moesin regulatory cytoskeletal proteins, consistent with a barrier-protective mechanism. Collectively, our data indicate that targeting adenosine receptors and downstream barrier-protective pathways in VVEC may have a potential translational significance in developing pharmacological approach for the VV barrier protection in PH.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Vasa Vasorum/metabolism , rac1 GTP-Binding Protein/metabolism , Adenosine/pharmacology , Animals , Cattle , Endothelial Cells/drug effects , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Male , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Vasa Vasorum/drug effects
2.
Am J Physiol Cell Physiol ; 312(1): C56-C70, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27856430

ABSTRACT

Angiogenesis is an energy-demanding process; however, the role of cellular energy pathways and their regulation by extracellular stimuli, especially extracellular nucleotides, remain largely unexplored. Using metabolic inhibitors of glycolysis (2-deoxyglucose) and oxidative phosphorylation (OXPHOS) (oligomycin, rotenone, and FCCP), we demonstrate that glycolysis and OXPHOS are both essential for angiogenic responses of vasa vasorum endothelial cell (VVEC). Treatment with P2R agonists, ATP, and 2-methylthioadenosine diphosphate trisodium salt (MeSADP), but not P1 receptor agonist, adenosine, increased glycolytic activity in VVEC (measured by extracellular acidification rate and lactate production). Stimulation of glycolysis was accompanied by increased levels of phospho-phosphofructokinase B3, hexokinase (HK), and GLUT-1, but not lactate dehydrogenase. Moreover, extracellular ATP and MeSADP, and to a lesser extent adenosine, increased basal and maximal oxygen consumption rates in VVEC. These effects were potentiated when the cells were cultured in 20 mM galactose and 5 mM glucose compared with 25 mM glucose. Treatment with P2R agonists decreased phosphorylation of pyruvate dehydrogenase (PDH)-E1α and increased succinate dehydrogenase (SDH), cytochrome oxidase IV, and ß-subunit of F1F0 ATP synthase expression. In addition, P2R stimulation transiently elevated mitochondrial Ca2+ concentration, implying involvement of mitochondria in VVEC angiogenic activation. We also demonstrated a critical role of phosphatidylinositol 3-kinase and Akt pathways in lactate production, PDH-E1α phosphorylation, and the expression of HK, SDH, and GLUT-1 in ATP-stimulated VVEC. Together, our findings suggest that purinergic and metabolic regulation of VVEC energy pathways is essential for VV angiogenesis and may contribute to pathologic vascular remodeling in pulmonary hypertension.


Subject(s)
Endothelial Cells/physiology , Glycolysis/physiology , Neovascularization, Physiologic/physiology , Oxidative Phosphorylation , Vasa Vasorum/cytology , Vasa Vasorum/physiology , Animals , Cattle , Cells, Cultured , Endothelial Cells/cytology , Male , Receptors, Purinergic
3.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L661-71, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24508729

ABSTRACT

Angiogenic expansion of the vasa vasorum (VV) is an important contributor to pulmonary vascular remodeling in the pathogenesis of pulmonary hypertension (PH). High proliferative potential endothelial progenitor-like cells have been described in vascular remodeling and angiogenesis in both systemic and pulmonary circulations. However, their role in hypoxia-induced pulmonary artery (PA) VV expansion in PH is not known. We hypothesized that profound PA VV neovascularization observed in a neonatal calf model of hypoxia-induced PH is due to increased numbers of subsets of high proliferative cells within the PA adventitial VV endothelial cells (VVEC). Using a single cell clonogenic assay, we found that high proliferative potential colony-forming cells (HPP-CFC) comprise a markedly higher percentage in VVEC populations isolated from the PA of hypoxic (VVEC-Hx) compared with control (VVEC-Co) calves. VVEC-Hx populations that comprised higher numbers of HPP-CFC also demonstrated markedly higher expression levels of CD31, CD105, and c-kit than VVEC-Co. In addition, significantly higher expression of CD31, CD105, and c-kit was observed in HPP-CFC vs. the VVEC of the control but not of hypoxic animals. HPP-CFC exhibited migratory and tube formation capabilities, two important attributes of angiogenic phenotype. Furthermore, HPP-CFC-Co and some HPP-CFC-Hx exhibited elevated telomerase activity, consistent with their high replicative potential, whereas a number of HPP-CFC-Hx exhibited impaired telomerase activity, suggestive of their senescence state. In conclusion, our data suggest that hypoxia-induced VV expansion involves an emergence of HPP-CFC populations of a distinct phenotype with increased angiogenic capabilities. These cells may serve as a potential target for regulating VVEC neovascularization.


Subject(s)
Hypertension, Pulmonary/etiology , Hypoxia/physiopathology , Neovascularization, Pathologic/pathology , Pulmonary Artery/pathology , Vasa Vasorum/physiopathology , Animals , Animals, Newborn , Antigens, CD/metabolism , Cattle , Cell Migration Assays , Cell Proliferation , Colony-Forming Units Assay , Hypertension, Pulmonary/physiopathology , Hypoxia/metabolism , Male , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Telomerase/metabolism
4.
PLoS One ; 8(4): e59733, 2013.
Article in English | MEDLINE | ID: mdl-23613714

ABSTRACT

BACKGROUND: In a neonatal model of hypoxic pulmonary hypertension, a dramatic pulmonary artery adventitial thickening, accumulation of inflammatory cells in the adventitial compartment, and angiogenic expansion of the vasa vasorum microcirculatory network are observed. These pathophysiological responses suggest that rapidly proliferating vasa vasorum endothelial cells (VVEC) may exhibit increased permeability for circulating blood cells and macromolecules. However, the molecular mechanisms underlying these observations remain unexplored. Some reports implicated extracellular adenosine in the regulation of vascular permeability under hypoxic and inflammatory conditions. Thus, we aimed to determine the role of adenosine in barrier regulation of VVEC isolated from the pulmonary arteries of normoxic (VVEC-Co) or chronically hypoxic (VVEC-Hyp) neonatal calves. PRINCIPAL FINDINGS: We demonstrate via a transendothelial electrical resistance measurement that exogenous adenosine significantly enhanced the barrier function in VVEC-Co and, to a lesser extent, in VVEC-Hyp. Our data from a quantitative reverse transcription polymerase chain reaction show that both VVEC-Co and VVEC-Hyp express all four adenosine receptors (A1, A2A, A2B, and A3), with the highest expression level of A1 receptors (A1Rs). However, A1R expression was significantly lower in VVEC-Hyp compared to VVEC-Co. By using an A1R-specific agonist/antagonist and siRNA, we demonstrate that A1Rs are mostly responsible for adenosine-induced enhancement in barrier function. Adenosine-induced barrier integrity enhancement was attenuated by pretreatment of VVEC with pertussis toxin and GSK690693 or LY294002, suggesting the involvement of Gi proteins and the PI3K-Akt pathway. Moreover, we reveal a critical role of actin cytoskeleton in VVEC barrier regulation by using specific inhibitors of actin and microtubule polymerization. Further, we show that adenosine pretreatment blocked the tumor necrosis factor alpha (TNF-α)-induced permeability in VVEC-Co, validating its anti-inflammatory effects. CONCLUSIONS: We demonstrate for the first time that stimulation of A1Rs enhances the barrier function in VVEC by activation of the Gi/PI3K/Akt pathway and remodeling of actin microfilament.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Adenosine A1/metabolism , Vasa Vasorum/cytology , Actin Cytoskeleton/drug effects , Animals , Cattle , Chromones/pharmacology , Endothelial Cells/drug effects , Male , Morpholines/pharmacology , Oxadiazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vasa Vasorum/drug effects , Vasa Vasorum/metabolism
5.
Annu Rev Physiol ; 75: 23-47, 2013.
Article in English | MEDLINE | ID: mdl-23216413

ABSTRACT

The vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and is composed of a variety of cells, including fibroblasts, immunomodulatory cells (dendritic cells and macrophages), progenitor cells, vasa vasorum endothelial cells and pericytes, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to influence the tone and structure of the vessel wall; to initiate and perpetuate chronic vascular inflammation; and to stimulate expansion of the vasa vasorum, which can act as a conduit for continued inflammatory and progenitor cell delivery to the vessel wall. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of vascular wall function and structure from the outside in.


Subject(s)
Adventitia/physiology , Blood Vessels/cytology , Blood Vessels/physiology , Adventitia/cytology , Animals , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Macrophages/cytology , Macrophages/physiology , Stem Cells/cytology , Stem Cells/physiology , Stress, Physiological/physiology , Vasa Vasorum/cytology , Vasa Vasorum/physiology
6.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L1-L11, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22582113

ABSTRACT

Increased cell proliferation and migration, of several cell types are key components of vascular remodeling observed in pulmonary hypertension (PH). Our previous data demonstrate that adventitial fibroblasts isolated from pulmonary arteries of chronically hypoxic hypertensive calves (termed PH-Fibs) exhibit a "constitutively activated" phenotype characterized by high proliferative and migratory potential. Osteopontin (OPN) has been shown to promote several cellular activities including growth and migration in cancer cells. We thus tested the hypothesis that elevated OPN expression confers the "activated" highly proproliferative and promigratory/invasive phenotype of PH-Fibs. Our results demonstrate that, both in vivo and ex vivo, PH-Fibs exhibited increased expression of OPN, as well as its cognate receptors, α(V)ß(3) and CD44, compared with control fibroblasts (CO-Fibs). Augmented OPN expression in PH-Fibs corresponded to their high proliferative, migratory, and invasive properties and constitutive activation of ERK1/2 and AKT signaling. OPN silencing via small interfering RNA or sequestering OPN production by specific antibodies led to decreased proliferation, migration, invasion, and attenuated ERK1/2, AKT phosphorylation in PH-Fibs. Furthermore, increasing OPN levels in CO-Fibs via recombinant OPN resulted in significant increases in their proliferative, migratory, and invasive capabilities to the levels resembling those of PH-Fibs. Thus our data suggest OPN as an essential contributor to the activated (highly proliferative, migratory, and proinvasive) phenotype of pulmonary adventitial fibroblasts in hypoxic PH.


Subject(s)
Fibroblasts/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Osteopontin/metabolism , Pulmonary Artery/metabolism , Animals , Cattle , Cell Growth Processes/physiology , Cell Hypoxia/physiology , Cell Movement/physiology , Cells, Cultured , Fibroblasts/pathology , Humans , Hyaluronan Receptors/metabolism , Hydrogen-Ion Concentration , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/pathology , Hypoxia/physiopathology , Integrin alphaVbeta3/metabolism , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System/physiology , Male , Neoplasm Invasiveness , Osteopontin/blood , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/pathology , Signal Transduction
7.
Angiogenesis ; 14(4): 503-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21922294

ABSTRACT

Vascular remodeling plays a pivotal role in a variety of pathophysiological conditions where hypoxia and inflammation are prominent features. Intravascular ATP, ADP and adenosine are known as important regulators of vascular tone, permeability and homeostasis, however contribution of purinergic signalling to endothelial cell growth and angiogenesis remains poorly understood. By using vasa vasorum endothelial cells (VVEC) isolated from pulmonary artery adventitia of control and chronically hypoxic neonatal calves, these studies were aimed to evaluate the effect of hypoxia on biochemical and functional properties of microvascular endothelial network at the sites of angiogenesis. In comparison with normoxic controls, VVEC from hypoxic animals are characterized by (1) drastically impaired nucleoside triphosphate diphosphohydrolase-1 (NTPDase-1/CD39) and ecto-5'-nucleotidase/CD73 activities with respective increases in basal extracellular ATP and ADP levels (2) higher proliferative responses to low micromolar concentrations of ATP and ADP; and (3) enhanced permeability and disordered adenosinergic control of vascular barrier function (measured as a paracellular flux of 70 kDa fluorescein isothiocyanate-dextran). Together, these results suggest that unique pattern of purine-mediated angiogenic activation and enhanced leakiness of VVEC from chronically hypoxic vessels may be defined by disordered endothelial nucleotide homeostasis at sites of active neovascularization.


Subject(s)
5'-Nucleotidase/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Endothelial Cells/metabolism , Hypoxia/metabolism , Neovascularization, Pathologic/metabolism , Pulmonary Artery/cytology , Vasa Vasorum/cytology , Adenosine/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Animals, Newborn , Blotting, Western , Capillary Permeability/physiology , Cattle , Cell Proliferation , Cyclic AMP/metabolism , DNA Primers/genetics , Dextrans , Fluorescein-5-isothiocyanate/analogs & derivatives , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction
8.
Vasc Cell ; 3(1): 4, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21349161

ABSTRACT

BACKGROUND: Platelets contribute to vascular homeostasis and angiogenesis through the release of multiple growth factors, cytokines and nucleotides, such as ATP and ADP. Recent reports have demonstrated a marked growth-promoting effect of total platelet extracts and selected platelet growth factors on therapeutic angiogenesis. However, since endogenous adenine nucleotides are rapidly degraded during the platelet isolation and storage, we examined whether supplementing a platelet-derived extract with exogenous adenine nucleotides would augment their pro-angiogenic effects. METHODS: Pulmonary artery vasa vasorum endothelial cells (VVEC) were used to examine the effects of dialyzed platelet-derived soluble extracts and extracellular adenine nucleotides on proliferation, migration and tube formation. In addition, an in vivo Matrigel plug assay was used to examine the effects of platelet extracts and adenine nucleotides on neovascularization of plugs subcutaneously placed in 50 ICR mice. The number of vascular structures in Matrigel plugs were evaluated by histological and statistical methods. RESULTS: Platelet extracts (6.4-64 µg/ml) significantly induced DNA synthesis and at a concentration of 64 µg/ml had a biphasic effect on VVEC proliferation (an increase at 48 hrs followed by a decrease at 60 hrs). Stimulation of VVEC with platelet extracts also significantly (up to several-fold) increased cell migration and tube formation on Matrigel. Stimulation of VVEC with extracellular ATP (100 µM) dramatically (up to ten-fold) increased migration and tube formation on Matrigel; however, no significant effects on cell proliferation were observed. We also found that ATP moderately diminished platelet extract-induced VVEC proliferation (48 hrs) and migration, but potentiated tube formation. Neither ATP, or a mixture of non-hydrolyzable nucleotides (ATPγS, ADPßS, MeSATP, MeSADP) induced vascularization of Matrigel plugs subcutaneously injected in mice, however, the combination of these nucleotides with platelet extracts dramatically increased the number of functional capillaries in the Matrigel plugs. CONCLUSION: Data from this study suggest that platelet-derived growth factors and extracellular nucleotides represent important regulatory signals for angiogenesis. Supplementation of platelet extracts with exogenous adenine nucleotides may reveal new possibilities for therapeutic angiogenesis and tissue regeneration approaches.

9.
Am J Physiol Cell Physiol ; 300(2): C266-75, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20962269

ABSTRACT

Extracellular ATP and ADP have been shown to exhibit potent angiogenic effects on pulmonary artery adventitial vasa vasorum endothelial cells (VVEC). However, the molecular signaling mechanisms of extracellular nucleotide-mediated angiogenesis remain not fully elucidated. Since elevation of intracellular Ca(2+) concentration ([Ca(2+)](i)) is required for cell proliferation and occurs in response to extracellular nucleotides, this study was undertaken to delineate the purinergic receptor subtypes involved in Ca(2+) signaling and extracellular nucleotide-mediated mitogenic responses in VVEC. Our data indicate that stimulation of VVEC with extracellular ATP resulted in the elevation of [Ca(2+)](i) via Ca(2+) influx through plasma membrane channels as well as Ca(2+) mobilization from intracellular stores. Moreover, extracellular ATP induced simultaneous Ca(2+) responses in both cytosolic and nuclear compartments. An increase in [Ca(2+)](i) was observed in response to a wide range of purinergic receptor agonists, including ATP, ADP, ATPγS, ADPßS, UTP, UDP, 2-methylthio-ATP (MeSATP), 2-methylthio-ADP (MeSADP), and BzATP, but not adenosine, AMP, diadenosine tetraphosphate, αßMeATP, and ßγMeATP. Using RT-PCR, we identified mRNA for the P2Y1, P2Y2, P2Y4, P2Y13, P2Y14, P2X2, P2X5, P2X7, A1, A2b, and A3 purinergic receptors in VVEC. Preincubation of VVEC with the P2Y1 selective antagonist MRS2179 and the P2Y13 selective antagonist MRS2211, as well as with pertussis toxin, attenuated at varying degrees agonist-induced intracellular Ca(2+) responses and activation of ERK1/2, Akt, and S6 ribosomal protein, indicating that P2Y1 and P2Y13 receptors play a major role in VVEC growth responses. Considering the broad physiological implications of purinergic signaling in the regulation of angiogenesis and vascular homeostasis, our findings suggest that P2Y1 and P2Y13 receptors may represent novel and specific targets for treatment of pathological vascular remodeling involving vasa vasorum expansion.


Subject(s)
Calcium Signaling , Calcium/physiology , Endothelium, Vascular/physiology , Pulmonary Artery/physiology , Receptors, Purinergic P2Y1/physiology , Vasa Vasorum/physiology , Adenosine Diphosphate/administration & dosage , Adenosine Diphosphate/analogs & derivatives , Animals , Azo Compounds/administration & dosage , Calcium/analysis , Calcium Channels/drug effects , Calcium Channels/physiology , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Endothelium, Vascular/drug effects , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/drug effects , Purinergic Agonists/metabolism , Pyridoxal Phosphate/administration & dosage , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic/drug effects , Receptors, Purinergic/physiology , Ribosomal Protein S6/metabolism , Vasa Vasorum/drug effects
10.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L954-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19684203

ABSTRACT

We recently reported that vasa vasorum expansion occurs in the pulmonary artery (PA) adventitia of chronically hypoxic animals and that extracellular ATP is a pro-angiogenic factor for isolated vasa vasorum endothelial cells (VVEC). However, the sources of extracellular ATP in the PA vascular wall, as well as the molecular mechanisms underlying its release, remain elusive. Studies were undertaken to explore whether VVEC release ATP in response to hypoxia and to determine signaling pathways involved in this process. We found that hypoxia (1-3% O2) resulted in time- and O2-dependent ATP release from VVEC. Preincubation with the inhibitors of vesicular transport (monensin, brefeldin A, and N-ethylmaleimide) significantly decreased ATP accumulation in the VVEC conditioned media, suggesting that hypoxia-induced ATP release occurs through vesicular exocytosis. Additionally, both hypoxia and exogenously added ATP resulted in the activation of PI3K and accumulation of GTP-bound RhoA in a time-dependent manner. Pharmacological inhibition of PI3K and ROCK or knockout of RhoA by small interfering RNA significantly abolished hypoxia-induced ATP release from VVEC. Moreover, RhoA and ROCK play a critical role in ATP-induced increases in VVEC DNA synthesis, migration, and tube formation, indicating a functional contribution of PI3K, Rho, and ROCK to both the autocrine mechanism of ATP release and ATP-mediated angiogenic activation of VVEC. Taken together, our findings provide novel evidence for the signaling mechanisms that link hypoxia-induced increases in extracellular ATP and vasa vasorum expansion.


Subject(s)
Adenosine Triphosphate/pharmacology , Endothelial Cells/enzymology , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Vasa Vasorum/cytology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Hypoxia/drug effects , Cell Movement/drug effects , Collagen/metabolism , DNA/biosynthesis , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Laminin/metabolism , Male , Proteoglycans/metabolism , Pulmonary Artery/cytology , Signal Transduction/drug effects , Time Factors , Transport Vesicles/drug effects , Transport Vesicles/metabolism
11.
Angiogenesis ; 11(2): 169-82, 2008.
Article in English | MEDLINE | ID: mdl-18071915

ABSTRACT

Expansion of the vasa vasorum network has been observed in a variety of systemic and pulmonary vascular diseases. We recently reported that a marked expansion of the vasa vasorum network occurs in the pulmonary artery adventitia of chronically hypoxic calves. Since hypoxia has been shown to stimulate ATP release from both vascular resident as well as circulatory blood cells, these studies were undertaken to determine if extracellular ATP exerts angiogenic effects on isolated vasa vasorum endothelial cells (VVEC) and/or if it augments the effects of other angiogenic factors (VEGF and basic FGF) known to be present in the hypoxic microenvironment. We found that extracellular ATP dramatically increases DNA synthesis, migration, and rearrangement into tube-like networks on Matrigel in VVEC, but not in pulmonary artery (MPAEC) or aortic (AOEC) endothelial cells obtained from the same animals. Extracellular ATP potentiated the effects of both VEGF and bFGF to stimulate DNA synthesis in VVEC but not in MPAEC and AOEC. Analysis of purine and pyrimidine nucleotides revealed that ATP, ADP and MeSADP were the most potent in stimulating mitogenic responses in VVEC, indicating the involvement of the family of P2Y1-like purinergic receptors. Using pharmacological inhibitors, Western blot analysis, and Phosphatidylinositol-3 kinase (PI3K) in vitro kinase assays, we found that PI3K/Akt/mTOR and ERK1/2 play a critical role in mediating the extracellular ATP-induced mitogenic and migratory responses in VVEC. However, PI3K/Akt and mTOR/p70S6K do not significantly contribute to extracellular ATP-induced tube formation on Matrigel. Our studies indicate that VVEC, isolated from the sites of active angiogenesis, exhibit distinct functional responses to ATP, compared to endothelial cells derived from large pulmonary or systemic vessels. Collectively, our data support the idea that extracellular ATP participates in the expansion of the vasa vasorum that can be observed in hypoxic conditions.


Subject(s)
Adenosine Triphosphate/pharmacology , Angiogenesis Inducing Agents/pharmacology , Endothelial Cells/drug effects , Extracellular Space/metabolism , Pulmonary Artery/cytology , Vasa Vasorum/cytology , Vasa Vasorum/drug effects , Animals , Aorta/cytology , Aorta/enzymology , Cattle , Cell Movement/drug effects , Collagen/metabolism , DNA/biosynthesis , Drug Combinations , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Space/drug effects , Fibroblast Growth Factor 2/pharmacology , Laminin/metabolism , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proteoglycans/metabolism , Pulmonary Artery/enzymology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Vasa Vasorum/enzymology , Vascular Endothelial Growth Factor A/pharmacology
12.
Am J Pathol ; 168(6): 1793-807, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16723696

ABSTRACT

The precise cellular and molecular mechanisms regulating adventitial vasa vasorum neovascularization, which occurs in the pulmonary arterial circulation in response to hypoxia, remain unknown. Here, using a technique to isolate and culture adventitial fibroblasts (AdvFBs) and vasa vasorum endothelial cells (VVECs) from the adventitia of pulmonary arteries, we report that hypoxia-activated pulmonary artery AdvFBs exhibited pro-angiogenic properties and influenced the angiogenic phenotype of VVEC, in a process of cell-cell communication involving endothelin-1 (ET-1). We demonstrated that AdvFBs, either via co-culture or conditioned media, stimulated VVEC proliferation and augmented the self-assembly and integrity of cord-like networks that formed when VVECs where cultured on Matrigel. In addition, hypoxia-activated AdvFBs produced ET-1, suggesting a paracrine role for this pro-angiogenic molecule in these processes. When co-cultured on Matrigel, AdvFBs and VVECs self-assembled into heterotypic cord-like networks, a process augmented by hypoxia but attenuated by either selective endothelin receptor antagonists or oligonucleotides targeting prepro-ET-1 mRNA. From these observations, we propose that hypoxia-activated AdvFBs exhibit pro-angiogenic properties and, as such, communicate with VVECs, in a process involving ET-1, to regulate vasa vasorum neovascularization occurring in the adventitia of pulmonary arteries in response to chronic hypoxia.


Subject(s)
Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/pathology , Hypoxia , Pulmonary Artery/metabolism , Animals , Cattle , Cell Culture Techniques/methods , Cell Proliferation , Cells, Cultured/metabolism , Disease Models, Animal , Hypertension/pathology , Microscopy, Fluorescence
14.
J Appl Physiol (1985) ; 98(2): 732-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15475598

ABSTRACT

In most mammalian species, chronic exposure to hypoxia leads to pulmonary hypertension and vascular remodeling. The adventitial fibroblast, because of its ability to proliferate in response to hypoxia, is thought to be a critical cell in the remodeling process. However, the transcription factors driving hypoxia-induced fibroblast proliferation have yet to be elucidated. The early growth response-1 (Egr-1) transcription factor has been shown to be upregulated by hypoxia in pulmonary artery adventitial fibroblasts. We therefore hypothesized that Egr-1 is directly involved in hypoxia-induced adventitial fibroblast proliferation. Immunohistochemical analysis of in vivo lung tissue from animals exposed to chronic hypoxia revealed increased expression of Egr-1 in the pulmonary artery fibroblasts vs. expression shown in normoxic controls. In fibroblasts cultured from chronically hypoxic animals, exposure to 1% oxygen upregulated Egr-1 protein and cell proliferation. To evaluate the role of Egr-1 in hypoxia-induced proliferation, we employed an Egr-1 antisense strategy. Addition of antisense Egr-1 oligonucleotides, but not sense oligonucleotides, attenuated the hypoxia-induced upregulation of Egr-1 protein and reduced hypoxia-induced DNA synthesis by 50%. Cell proliferation was also significantly inhibited by the addition of antisense Egr-1 oligonucleotides but not the sense oligonucleotides. In addition, hypoxia-induced upregulations of cyclin D and epidermal growth factor receptor were attenuated by Egr-1 antisense oligonucleotides. We conclude that Egr-1 protein expression is very sensitive to upregulation by hypoxia in pulmonary artery adventitial fibroblasts and that it plays an important role in the autonomous growth phenotype induced by hypoxia in these cells.


Subject(s)
Cell Hypoxia/physiology , Fibroblasts/physiology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Pulmonary Artery/growth & development , Pulmonary Artery/metabolism , Animals , Cattle , Cell Proliferation , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Gene Silencing , Oligonucleotides, Antisense/administration & dosage , Pulmonary Artery/cytology , Pulmonary Artery/embryology , Signal Transduction/genetics
15.
J Appl Physiol (1985) ; 98(2): 722-31, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15501927

ABSTRACT

In contrast to cell types in which exposure to hypoxia causes a general reduction of metabolic activity, a remarkable feature of pulmonary artery adventitial fibroblasts is their ability to proliferate in response to hypoxia. Previous studies have suggested that ERK1/2, phosphatidylinositol 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR) are activated by hypoxia and play a role in a variety of cell responses. However, the pathways involved in mediating hypoxia-induced proliferation are largely unknown. Using pharmacological inhibitors, we established that PI3K-Akt, mTOR-p70 ribosomal protein S6 kinase (p70S6K), and EKR1/2 signaling pathways play a critical role in hypoxia-induced adventitial fibroblast proliferation. We found that exposure of serum-starved fibroblasts to 3% O2 resulted in a time-dependent activation of PI3K and transient phosphorylation of Akt. However, activation of PI3K was not required for activation of ERK1/2, implying a parallel involvement of these pathways in the proliferative response of fibroblasts to hypoxia. We found that hypoxia induced significant increases in mTOR, p70S6K, 4E-BP1, and S6 ribosomal protein phosphorylation, as well as dramatic increases in p70S6K activity. The activation of p70S6K/S6 pathway was sensitive to inhibition by rapamycin and LY294002, indicating that mTOR and PI3K/Akt are upstream signaling regulators. However, the magnitude of hypoxia-induced p70S6K activity and phosphorylation suggests involvement of additional signaling pathways. Thus our data demonstrate that hypoxia-induced adventitial fibroblast proliferation requires activation and interaction of PI3K, Akt, mTOR, p70S6K, and ERK1/2 and provide evidence for hypoxic regulation of protein translational pathways in cells exhibiting the capability to proliferate under hypoxic conditions.


Subject(s)
Cell Hypoxia/physiology , Fibroblasts/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pulmonary Artery/growth & development , Pulmonary Artery/metabolism , Animals , Cattle , Cell Proliferation , Cells, Cultured , Enzyme Activation , Fibroblasts/cytology , Proto-Oncogene Proteins c-akt , Pulmonary Artery/cytology , Pulmonary Artery/embryology , Signal Transduction/physiology , TOR Serine-Threonine Kinases
16.
J Biol Chem ; 280(3): 1838-48, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15522879

ABSTRACT

Extracellular nucleotides are increasingly recognized as important regulators of growth in a variety of cell types. Recent studies have demonstrated that extracellular ATP is a potent inducer of fibroblast growth acting, at least in part, through an ERK1/2-dependent signaling pathway. However, the contributions of additional signaling pathways to extracellular ATP-mediated cell proliferation have not been defined. By using both pharmacologic and genetic approaches, we found that in addition to ERK1/2, phosphatidylinositol 3-kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), and p70 S6K-dependent signaling pathways are required for ATP-induced proliferation of adventitial fibroblasts. We found that extracellular ATP acting in part through G(i) proteins increased PI3K activity in a time-dependent manner and transient phosphorylation of Akt. This PI3K pathway is not involved in ATP-induced activation of ERK1/2, implying activation of independent parallel signaling pathways by ATP. Extracellular ATP induced dramatic increases in mTOR and p70 S6K phosphorylation. This activation of the mTOR/p70 S6 kinase (p70 S6K) pathway in response to ATP is because of independent contributions of PI3K/Akt and ERK1/2 pathways, which converge on the level of p70 S6K. ATP-dependent activation of mTOR and p70 S6K also requires additional signaling inputs perhaps from pathways operating through Galpha or Gbetagamma subunits. Collectively, our data demonstrate that ATP-induced adventitial fibroblast proliferation requires activation and interaction of multiple signaling pathways such as PI3K, Akt, mTOR, p70 S6K, and ERK1/2 and provide evidence for purinergic regulation of the protein translational pathways related to cell proliferation.


Subject(s)
Adenosine Triphosphate/physiology , Cell Division/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Animals , Cattle , Cells, Cultured , Proto-Oncogene Proteins c-akt , Pulmonary Artery/cytology , Pulmonary Artery/enzymology , Pulmonary Artery/metabolism , TOR Serine-Threonine Kinases
17.
J Biol Chem ; 277(47): 44638-50, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12244041

ABSTRACT

Important autocrine/paracrine functions for the adenine nucleotides have been proposed in several tissues. We addressed the possibility that extracellular ATP would modulate/mediate hypoxia-induced adventitial fibroblast growth. Acute hypoxia (3% O(2), 10-60 min) increased extracellular ATP concentrations in adventitial fibroblasts and in lung microvascular endothelial cells, and chronic hypoxia (3% O(2), 14-30 days) markedly attenuated the rate of extracellular ATP hydrolysis by ecto-nucleotidase(s). Exogenous ATP stimulated [(3)H]thymidine incorporation in fibroblasts as did UTP, ADPbeta, 2-methylthioadenosine triphosphate, adenosine 5'-(alpha,beta-methylene)triphosphate, and benzoylbenzoyl-ATP (2'-3'-O-(4-benzoylbenzoyl)-ATP), indicating that both P2Y and P2X purinoceptors can mediate mitogenic responses. Suramin (100 microm), Cibacron blue 3GA (100 microm), and pyridoxalphosphate-6-azophenyl-2',-4'-disulfonic acid (100 microm) as well as apyrase (5 units/ml) attenuated hypoxia- and ATP-induced and DNA synthesis, indicating activation and a functional role of purinoceptors under hypoxic conditions. ATP-induced DNA synthesis was augmented by hypoxia in an additive fashion, whereas ATP and hypoxia synergistically increased growth factor-induced DNA synthesis, again suggesting that ATP and hypoxia utilize similar signaling pathways to induce proliferation. Indeed, we found that ATP (100 microm) and hypoxia (3% O(2)) induced expression and activation of Egr-1 transcription factor, and both stimuli acted, in part, through a G(alpha)(i)/ERK1/2-dependent signaling pathway. Suramin, Cibacron blue 3GA, and apyrase attenuated hypoxia-induced ERK1/2 activation and Egr-1 expression. We conclude that hypoxia induces ATP release from endothelial cells and fibroblasts and that the activation of P2 purinoceptors is involved in the regulation of DNA synthesis by fibroblasts under hypoxic conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Autocrine Communication/physiology , Cell Hypoxia , DNA-Binding Proteins/metabolism , Fibroblasts/physiology , Immediate-Early Proteins , Paracrine Communication/physiology , Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cattle , Cell Division/physiology , Cells, Cultured , Culture Media, Serum-Free , DNA/biosynthesis , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Early Growth Response Protein 1 , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fetus/anatomy & histology , Fibroblasts/cytology , Fibroblasts/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Growth Substances/pharmacology , Humans , Lung/cytology , Lung/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nucleotides/metabolism , Protein Binding , Purinergic P2 Receptor Antagonists , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL