Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
FASEB J ; 22(8): 3010-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18502941

ABSTRACT

Angiopoietins were thought to be endothelial cell-specific via the tie2 receptor. We showed that angiopoietin-1 (ang1) also interacts with integrins on cardiac myocytes (CMs) to increase survival. Because ang1 monomers bind and activate integrins (not tie2), we determined their function in vivo. We examined monomer and multimer expressions during physiological and pathological cardiac remodeling and overexpressed ang1 monomers in phenylephrine-induced cardiac hypertrophy. Cardiac ang1 levels (mRNA, protein) increased during postnatal development and decreased with phenylephrine-induced cardiac hypertrophy, whereas tie2 phosphorylations were unchanged. We found that most or all of the changes during cardiac remodeling were in monomers, offering an explanation for unchanged tie2 activity. Heart tissue contains abundant ang1 monomers and few multimers (Western blotting). We generated plasmids that produce ang1 monomers (ang1-256), injected them into mice, and confirmed cardiac expression (immunohistochemistry, RT-PCR). Ang1 monomers localize to CMs, smooth muscle cells, and endothelial cells. In phenylephrine-induced cardiac hypertrophy, ang1-256 reduced left ventricle (LV)/tibia ratios, fetal gene expressions (atrial and brain natriuretic peptides, skeletal actin, beta-myosin heavy chain), and fibrosis (collagen III), and increased LV prosurvival signaling (akt, MAPK(p42/44)), and AMPK(T172). However, tie2 phosphorylations were unchanged. Ang1-256 increased integrin-linked kinase, a key regulator of integrin signaling and cardiac health. Collectively, these results suggest a role for ang1 monomers in cardiac remodeling.


Subject(s)
Angiopoietin-1/chemistry , Angiopoietin-1/metabolism , Cardiomegaly/prevention & control , Integrins/metabolism , Angiopoietin-1/genetics , Animals , Base Sequence , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cell Line , DNA Primers/genetics , Endothelial Cells/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phenylephrine/toxicity , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, TIE-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
2.
Circ Res ; 101(3): 286-94, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17569886

ABSTRACT

We report that human embryonic stem cells contain a population of vascular progenitor cells that have the ability to differentiate into endothelial-like and smooth muscle (SM)-like cells. Vascular progenitor cells were isolated from EBs grown in suspension for 10 days and were characterized by expression of the endothelial/hematopoietic marker CD34 (CD34+ cells). When these cells are subsequently cultured in EGM-2 (endothelial growth medium) supplemented with vascular endothelial growth factor-165 (50 ng/mL), they give rise to endothelial-like cells characterized by a cobblestone cell morphology, expression of endothelial markers (platelet endothelial cell-adhesion molecule-1, CD34, KDR/Flk-1, vascular endothelial cadherin, von Willebrand factor), incorporation of acetylated low-density lipoprotein, and formation of capillary-like structures when placed in Matrigel. In contrast, when CD34+ cells are cultured in EGM-2 supplemented with platelet-derived growth factor-BB (50 ng/mL), they give rise to SM-like cells characterized by spindle-shape morphology, expression of SM cell markers (alpha-SM actin, SM myosin heavy chain, calponin, caldesmon, SM alpha-22), and the ability to contract and relax in response to common pharmacological agents such as carbachol and atropine but rarely form capillary-like structures when placed in Matrigel. Implantation studies in nude mice show that both cell types contribute to the formation of human microvasculature. Some microvessels contained mouse blood cells, which indicates functional integration with host vasculature. Therefore, the vascular progenitors isolated from human embryonic stem cells using methods established in the present study could provide a means to examine the mechanisms of endothelial and SM cell development, and they could also provide a potential source of cells for vascular tissue engineering.


Subject(s)
Blood Vessels/cytology , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Myocytes, Smooth Muscle/cytology , Animals , Antigens, CD34/biosynthesis , Becaplermin , Biomarkers , Blood Vessels/ultrastructure , Cattle , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/transplantation , Collagen , Culture Media/pharmacology , Drug Combinations , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Fetal Blood , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Injections, Subcutaneous , Laminin , Mice , Mice, Nude , Morphogenesis , Muscle Proteins/biosynthesis , Neovascularization, Physiologic , Organ Specificity , Platelet-Derived Growth Factor/pharmacology , Proteoglycans , Proto-Oncogene Proteins c-sis , Tissue Engineering , Vascular Endothelial Growth Factor A/pharmacology
3.
Lancet ; 370(9604): 2011-9, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18083403

ABSTRACT

BACKGROUND: Sunitinib, a multitargeted tyrosine-kinase inhibitor, which is approved by both US and European Commission regulatory agencies for clinical use, extends survival of patients with metastatic renal-cell carcinoma and gastrointestinal stromal tumours, but concerns have arisen about its cardiac safety. We therefore assessed the cardiovascular risk associated with sunitinib in patients with metastatic gastrointestinal stromal tumours. METHODS: We retrospectively reviewed all cardiovascular events in 75 patients with imatinib-resistant, metastatic, gastrointestinal stromal tumours who had been enrolled in a phase I/II trial investigating the efficacy of sunitinib. The composite cardiovascular endpoint was cardiac death, myocardial infarction, and congestive heart failure. We also examined sunitinib's effects on left ventricular ejection fraction (LVEF) and blood pressure. We investigated potential mechanisms of sunitinib-associated cardiac effects by studies in isolated rat cardiomyocytes and in mice. FINDINGS: Eight of 75 (11%) patients given repeating cycles of sunitinib in the phase I/II trial had a cardiovascular event, with congestive heart failure recorded in six of 75 (8%). Ten of 36 (28%) patients treated at the approved sunitinib dose had absolute LVEF reductions in ejection fraction (EF) of at least 10%, and seven of 36 (19%) had LVEF reductions of 15 EF% or more. Sunitinib induced increases in mean systolic and diastolic blood pressure, and 35 of 75 (47%) individuals developed hypertension (>150/100 mm Hg). Congestive heart failure and left ventricular dysfunction generally responded to sunitinib being withheld and institution of medical management. Sunitinib caused mitochondrial injury and cardiomyocyte apoptosis in mice and in cultured rat cardiomyocytes. INTERPRETATION: Left ventricular dysfunction might be due, in part, to direct cardiomyocyte toxicity, exacerbated by hypertension. Patients treated with sunitinib should be closely monitored for hypertension and LVEF reduction, especially those with a history of coronary artery disease or cardiac risk factors.


Subject(s)
Antineoplastic Agents/adverse effects , Heart Failure/chemically induced , Indoles/adverse effects , Pyrroles/adverse effects , Animals , Antineoplastic Agents/therapeutic use , Blood Pressure/drug effects , Female , Gastrointestinal Stromal Tumors/drug therapy , Humans , Indoles/therapeutic use , Male , Mice , Middle Aged , Multicenter Studies as Topic , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/therapeutic use , Rats , Retrospective Studies , Stroke Volume/drug effects , Sunitinib
4.
Circ Res ; 96(4): e8-24, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15692086

ABSTRACT

Cardiac myocyte loss, regardless of insult, can trigger compensatory myocardial remodeling leading to heart failure. Identifying mediators of cardiac myocyte survival may advance clinical efforts toward myocardial preservation. Angiopoietin-1 limits ischemia-induced cardiac injury. This benefit is ascribed to angiogenesis because the receptor, tie2, is largely endothelial-specific. We propose that direct, non-tie2 interactions of angiopoietin-1 on cardiac myocytes contribute to this cardioprotection. We found that mouse C2C12 skeletal myocytes lack tie2, yet dose-dependently adhered to angiopoietin-1 and angiopoietin-2 similarly to laminin, fibronectin, vitronectin, and more than to collagen-I, -III, and -IV. Adhesion was divalent cation-mediated (Mn2+, Ca2+, not Mg2+), blocked with EDTA/EGTA, RGD-based peptides, and select integrin subunit antibodies. Similar findings were obtained with human skeletal myocytes (HSMs) and freshly isolated rat neonatal cardiac myocytes (NCMs). Furthermore, angiopoietin-1 conferred significant survival advantage exceeding that of most cell matrices, which was not fully explained by differences in cell adhesion. Angiopoietin-1 promoted survival of serum-starved C2C12, HSM, and NCM (MTT, trypan blue) and prevented taxol-induced apoptosis (caspase-3). Immobilized and soluble angiopoietin-1 phosphorylated Akt(S473) and MAPK(p42/44), (not FAK(Y397)) in C2C12 more than in endothelial cells and more than did angiopoietin-2 or cell matrices. EDTA, RGD-based peptides, and some integrin antibodies blocked these responses. Angiopoietin-1 activated HSM and NCM Akt(S473) and MAPK(p42/44) survival pathways. We propose that this novel function contributes to developmental and cardioprotective actions of angiopoietin-1 presently attributed to vascular effects alone. Angiopoietin-1 may prove therapeutically valuable in cardiac remodeling by supporting myocyte viability and preserving pump function. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Angiopoietin-1/pharmacology , Integrins/physiology , Muscle Fibers, Skeletal/drug effects , Myocytes, Cardiac/drug effects , Amino Acid Substitution , Angiopoietin-1/genetics , Angiopoietin-1/physiology , Angiopoietin-2/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Adhesion/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Edetic Acid/pharmacology , Extracellular Matrix Proteins/physiology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oligopeptides/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , Receptor, TIE-2/physiology , Recombinant Fusion Proteins/pharmacology
5.
Fertil Steril ; 83(1): 171-81, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15652904

ABSTRACT

OBJECTIVE: To determine whether nonsteroidal antiinflammatory drugs (NSAIDs) affect the establishment and progression of endometriotic lesions in a murine model. DESIGN: Pharmacologic intervention in a surgically induced murine model of abdominal/peritoneal endometriosis. SETTING: Animal research facility. PATIENT(S): Eight-week-old, female C57BL/6 mice. INTERVENTION(S): After implantation of autologous endometrium, mice were randomized into groups and treated with one of several NSAIDs or the vehicle-matched control for 4 weeks. MAIN OUTCOME MEASURE(S): Establishment, growth, and total burden of endometriotic lesions. RESULT(S): The NSAIDs differentially inhibited lesion establishment and growth, resulting in significantly reduced disease burden. Compared with controls (5.7 +/- 2.3 mm(2)), lesion burden was reduced by celecoxib (1.3 +/- 1.2 mm(2)), indomethacin (1.4 +/- 1.4 mm(2)), naproxen (2.7 +/- 1.2 mm(2)), sulindac (3.1 +/- 1.5 mm(2)), rofecoxib (3.4 +/- 3.0 mm(2)), and ibuprofen (4.1 +/- 1.4 mm(2)). In contrast, aspirin (5.9 +/- 1.2 mm(2)) had no statistically significant effect. Uninterrupted estrus cycling was confirmed by vaginal exams and smears in celecoxib-treated mice. CONCLUSION(S): Chronic administration of certain NSAIDs limits the progression of endometriosis in this murine model. The data suggest that NSAID selection in the treatment of endometriosis should be extended beyond pain management to maximize the inhibitory effect on disease burden.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Endometriosis/drug therapy , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Animals , Celecoxib , Disease Models, Animal , Endometriosis/pathology , Female , Humans , Mice , Mice, Inbred C57BL
6.
Hypertension ; 58(1): 85-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482957

ABSTRACT

Vascular endothelial growth factor regulates neoplastic angiogenesis through production of endothelium-derived NO. We performed a prospective evaluation of vascular function during treatment with vandetanib, a vascular endothelial growth receptor 2 and 3 receptor tyrosine kinase inhibitor, to determine the effects of vascular endothelial growth receptor signal interruption on endothelial function in humans. Seventeen patients with stage IV breast cancer received dose-escalated vandetanib in combination with low-dose oral chemotherapy. We measured blood pressure, systemic nitrate/nitrite levels, and brachial artery vascular function. In vitro analyses of cultured endothelial cells were performed to determine the effect of vandetanib on NO production, akt(473) phosphorylation, and endothelial NO synthase protein content and membrane localization. Vandetanib treatment for 6 weeks significantly increased blood pressure, decreased resting brachial artery diameter, and decreased plasma systemic nitrate/nitrite levels compared with baseline. Flow-mediated vasodilation was preserved, and no change was noted in nitroglycerin-mediated vasodilation. In vitro, endothelial cell nitrite levels and akt(473) phosphorylation were reduced and vascular endothelial growth receptor 2 levels did not change, but endothelial NO synthase membrane concentration doubled. Vandetanib reduces constitutive NO production and increases blood pressure, yet flow-stimulated NO bioavailability was preserved. Changes in vascular function with tyrosine kinase inhibition are complex and require further study in humans.


Subject(s)
Breast Neoplasms/drug therapy , Endothelium, Vascular/metabolism , Neovascularization, Pathologic/prevention & control , Nitric Oxide/biosynthesis , Piperidines/administration & dosage , Quinazolines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Vasodilation/drug effects , Administration, Oral , Adult , Aged , Blood Pressure/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Prospective Studies , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
7.
J Biomed Mater Res A ; 95(1): 105-17, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20540095

ABSTRACT

Myocardial infarction (MI) results in the death of cardiomyocytes (CM), which causes scar formation and pathological remodeling of the heart. The delivery of healthy myocytes or bone marrow cells reduces pathological remodeling after MI, however, current cell injection methods have low cell survival rates and high cell loss. The main objective of this work was to develop a novel hydrogel that can promote survival of CMs. Photocrosslinkable azidobenzoic acid modified chitosan (Az-chitosan) was conjugated with the angiopoietin-1-derived peptide, QHREDGS. This novel peptide is thought to mediate attachment and survival responses of CM to angiopoietin-1 via integrin binding. Thin layers of Az-chitosan, Az-chitosan-QHREDGS, and Az-chitosan-DGQESHR (scrambled peptide control) were spin coated on glass slides and photocrosslinked with application of UV light (365 nm). Neonatal rat heart cells cultured up to 5 days, demonstrated significantly higher attachment and viability on Az-chitosan-QHREDGS compared to cells on other hydrogel controls. Surfaces were also stained for the CM-specific marker troponin I, demonstrating significantly higher percentage of CMs on Az-chitosan-QHREDGS compared to Az-chitosan. The cells cultivated on Az-chitosan-QHREDGS demonstrated significantly lower levels of caspase 3/7 activation after taxol treatment in comparison to cells cultivated on the control hydrogels, glass substrate, or Az-chitosan linked to RGD, an established integrin binding peptide that did not protect against apoptosis. Thus, Az-chitosan-QHREDGS supports attachment and survival of neonatal rat heart cells.


Subject(s)
Angiopoietin-1/pharmacology , Chitosan/pharmacology , Cross-Linking Reagents/pharmacology , Myocardium/cytology , Myocytes, Cardiac/cytology , Peptides/pharmacology , Ultraviolet Rays , Amino Acid Sequence , Angiopoietin-1/chemistry , Animals , Animals, Newborn , Apoptosis/drug effects , Azides/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Microscopy, Electron, Scanning , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Peptides/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties/drug effects , Troponin I/metabolism
8.
Fertil Steril ; 84 Suppl 2: 1144-55, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16210006

ABSTRACT

OBJECTIVE: To determine whether endometriosis can be treated with the angiogenesis inhibitor endostatin and the effect of this treatment on fertility and reproduction. DESIGN: Pharmacologic intervention in a surgically induced model of endometriosis and in female mice undergoing mating. SETTING: Animal research facility. ANIMAL(S): Eight-week-old, female C57BL/6 and SCID mice. INTERVENTION(S): After implantation of autologous endometrium, mice received endostatin or the vehicle-matched control for 4 weeks. For the reproductive function study, mice receiving endostatin or vehicle were mated and reproductive functions were observed. MAIN OUTCOME MEASURE(S): Growth of endometriotic lesions after 4 weeks of treatment; estrous cycling, corpus luteum formation, serum hormone levels, and mating time as fertility measures; and pregnancy rates, length of pregnancy, fetal vitality, number, and outcome of litter as reproductive measures. RESULT(S): Endostatin suppressed the growth of endometriotic lesions by 47% compared with controls. Estrous cycling and corpus luteum formation were normal in both groups. Female mice receiving endostatin were as fertile as mice receiving vehicle, had normal pregnancies, and delivered the same number of pups. The offspring were healthy without teratogenic stigmata and reproduced normally themselves. CONCLUSION(S): Antiangiogenic therapy with endostatin may present a promising novel, nontoxic therapeutic option for patients with endometriosis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endometriosis/drug therapy , Endometriosis/pathology , Endostatins/therapeutic use , Fertility/drug effects , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Line, Tumor , Endostatins/administration & dosage , Female , Fertility/physiology , Growth Inhibitors/administration & dosage , Growth Inhibitors/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Pregnancy
9.
Proc Natl Acad Sci U S A ; 99(16): 10730-5, 2002 Aug 06.
Article in English | MEDLINE | ID: mdl-12149466

ABSTRACT

Tumor growth is angiogenesis dependent. We hypothesized that nonneoplastic tissue growth also depends on neovascularization. We chose adipose tissue as an experimental system because of its remodeling capacity. Mice from different obesity models received anti-angiogenic agents. Treatment resulted in dose-dependent, reversible weight reduction and adipose tissue loss. Marked vascular remodeling was evident in adipose tissue sections, which revealed decreased endothelial proliferation and increased apoptosis in treated mice compared with controls. Continuous treatment maintained mice near normal body weights for age without adverse effects. Metabolic adaptations in food intake, metabolic rate, and energy substrate utilization were associated with anti-angiogenic weight loss. We conclude that adipose tissue mass is sensitive to angiogenesis inhibitors and can be regulated by its vasculature.


Subject(s)
Adipose Tissue/blood supply , Angiogenesis Inhibitors/pharmacology , Neovascularization, Pathologic , Obesity/physiopathology , Organic Chemicals , Adipose Tissue/drug effects , Angiostatins , Animals , Antineoplastic Agents/pharmacology , Biphenyl Compounds , Body Composition/drug effects , Body Weight/drug effects , Collagen/pharmacology , Cyclohexanes , Disease Models, Animal , Endostatins , Energy Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , O-(Chloroacetylcarbamoyl)fumagillol , Obesity/metabolism , Peptide Fragments/pharmacology , Phenylbutyrates , Plasminogen/pharmacology , Sesquiterpenes/pharmacology , Thalidomide/pharmacology , Time Factors
10.
Biochem Biophys Res Commun ; 311(3): 563-71, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-14623307

ABSTRACT

Adipose tissue is unique in its plasticity, capacity for vascular remodeling, and susceptibility to angiogenesis inhibitors. We hypothesize that these characteristics are enabled by maintaining relatively immature adipose vessels to facilitate vascular/tissue remodeling. We examined the vascular maturation regulators, angiopoietin-1, angiopoietin-2, and tie2 receptor, under different weight-modifying conditions. Adipocytes expressed angiopoietin-1, while adipose endothelial cells expressed angiopoietin-2 and tie2. Adipose tissue growth/regression were associated with decreased angiopoietin-1 mRNA and protein, and tie2 phosphorylation. Angiopoietin-2 and tie2 mRNA levels were stable. Angiopoietin-1 mRNA levels inversely correlated with the rates of change in body weight, independent of the direction (weight gain, loss) or etiology (TNP-470, leptin, and diet restriction) of the weight shift. Obese mice injected with ang1/pcDNA had reduced rates of weight gain and fat pad weights, regardless of the route of plasmid administration (subcutaneous, intramuscular, and intravenous). Thus, angiopoietin-1 may regulate adipose tissue growth, suggesting that vascular maturation alters tissue plasticity.


Subject(s)
Adipose Tissue/physiology , Angiopoietin-1/physiology , 3T3-L1 Cells , Angiopoietin-2/biosynthesis , Animals , Blotting, Northern , Blotting, Western , Cyclohexanes , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , O-(Chloroacetylcarbamoyl)fumagillol , Plasmids/metabolism , Precipitin Tests , RNA, Messenger/metabolism , Receptor, TIE-2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sesquiterpenes/pharmacology , Time Factors , Tissue Distribution , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL