Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Z Evid Fortbild Qual Gesundhwes ; 129: 27-30, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29153351

ABSTRACT

Some medical diagnostic and therapeutic interventions are non-beneficial or even harmful. The Choosing Wisely campaign has encouraged the generation of "top five" lists of unnecessary low-value services in different specialist areas. In the USA alone, where the campaign was launched, these lists include a total of 450 evidence-based recommendations. Medical scientific societies in further countries such as Canada, Australia, New Zealand, England, Switzerland and Germany have since initiated Choosing Wisely campaigns. Besides implementing top five lists, these aim to change attitudes, expectations and practices in the culture of medicine. The field of internal medicine has initiated change in Switzerland (Swiss Society of General Internal Medicine: Smarter Medicine) and Germany (German Society of Internal Medicine: Klug entscheiden). Formulating Choosing Wisely principles in managing complex patients with multiple concurrent acute or chronic diseases, i. e., multimorbidity (MM), will present a particular challenge. Research is needed to determine the primary sources of overuse in specific combinations of diseases (i. e., MM clusters) and spearhead corresponding recommendations. National Choosing Widely campaigns may serve as a forerunner to a more global initiative.


Subject(s)
Clinical Competence , Multimorbidity , Societies, Medical , Australia , Canada , England , Germany , Humans , Societies, Medical/standards , Switzerland
2.
Sci Rep ; 5: 17705, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26635098

ABSTRACT

To explore the general requirement of endothelial mTORC2 during embryonic and adolescent development, we knocked out the essential mTORC2 component Rictor in the mouse endothelium in the embryo, during adolescence and in endothelial cells in vitro. During embryonic development, Rictor knockout resulted in growth retardation and lethality around embryonic day 12. We detected reduced peripheral vascularization and delayed ossification of developing fingers, toes and vertebrae during this confined midgestational period. Rictor knockout did not affect viability, weight gain, and vascular development during further adolescence. However during this period, Rictor knockout prevented skin capillaries to gain larger and heterogeneously sized diameters and remodeling into tortuous vessels in response to FGF2. Rictor knockout strongly reduced extensive FGF2-induced neovascularization and prevented hemorrhage in FGF2-loaded matrigel plugs. Rictor knockout also disabled the formation of capillary-like networks by FGF2-stimulated mouse aortic endothelial cells in vitro. Low RICTOR expression was detected in quiescent, confluent mouse aortic endothelial cells, whereas high doses of FGF2 induced high RICTOR expression that was associated with strong mTORC2-specific protein kinase Cα and AKT phosphorylation. We demonstrate that the endothelial FGF-RICTOR axis is not required during endothelial quiescence, but crucial for midgestational development and sustained and extensive neovascularization in the adult.


Subject(s)
Carrier Proteins/biosynthesis , Embryonic Development/genetics , Fibroblast Growth Factor 2/genetics , Neovascularization, Physiologic/genetics , Animals , Carrier Proteins/genetics , Endothelium/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Developmental , Hemorrhage/genetics , Hemorrhage/pathology , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-akt/genetics , Rapamycin-Insensitive Companion of mTOR Protein , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
Biochem Biophys Res Commun ; 461(2): 287-92, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25881506

ABSTRACT

Obesity involves hypoxic adipose tissue and low-grade chronic inflammation. We investigated the impact of hypoxia on inflammatory response to TNF-α in white and brown adipocytes. In response to TNF-α, the expression of the inducible enzymes iNOS and COX-2 was prominently and selectively potentiated during hypoxia while only moderately under normoxia. Levels of their products, nitrite and prostaglandinE2 were elevated accordingly. NS398, a selective COX-2 inhibitor, reduced nitrite levels. The expression of PGC-1α, a transcriptional co-activator involved in mitochondrial biogenesis, and PPARγ, a transcription factor involved in adipocyte homeostasis, was reduced by TNF-α during hypoxia. These results suggest that hypoxia potentiates the inflammatory response by TNF-α in both white and brown adipocytes and downregulates the transcription factors involved in adipocyte function.


Subject(s)
Adipocytes/immunology , Cyclooxygenase 2/genetics , Gene Expression Regulation , Hypoxia/immunology , Nitric Oxide Synthase Type II/genetics , Tumor Necrosis Factor-alpha/immunology , Adipocytes/metabolism , Adipocytes/pathology , Adipocytes, Brown/immunology , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Adipocytes, White/immunology , Adipocytes, White/metabolism , Adipocytes, White/pathology , Animals , Cell Hypoxia , Cell Line , Cyclooxygenase 2/analysis , Hypoxia/genetics , Hypoxia/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Nitric Oxide Synthase Type II/analysis
4.
Arterioscler Thromb Vasc Biol ; 33(9): 2105-11, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868942

ABSTRACT

OBJECTIVE: Perivascular adipose tissue (PVAT) wraps blood vessels and modulates vasoreactivity by secretion of vasoactive molecules. Mammalian target of rapamycin complex 2 (mTORC2) has been shown to control inflammation and is expressed in adipose tissue. In this study, we investigated whether adipose-specific deletion of rictor and thereby inactivation of mTORC2 in PVAT may modulate vascular function by increasing inflammation in PVAT. APPROACH AND RESULTS: Rictor, an essential mTORC2 component, was deleted specifically in mouse adipose tissue (rictor(ad-/-)). Phosphorylation of mTORC2 downstream target Akt at Serine 473 was reduced in PVAT from rictor(ad-/-) mice but unaffected in aortic tissue. Ex vivo functional analysis of thoracic aortae revealed increased contractions and impaired dilation in rings with PVAT from rictor(ad-/-) mice. Adipose rictor knockout increased gene expression and protein release of interleukin-6, macrophage inflammatory protein-1α, and tumor necrosis factor-α in PVAT as shown by quantitative real-time polymerase chain reaction and Bioplex analysis for the cytokines in the conditioned media, respectively. Moreover, gene and protein expression of inducible nitric oxide synthase was upregulated without affecting macrophage infiltration in PVAT from rictor(ad-/-) mice. Inhibition of inducible nitric oxide synthase normalized vascular reactivity in aortic rings from rictor(ad-/-) mice with no effect in rictor(fl/fl) mice. Interestingly, in perivascular and epididymal adipose depots, high-fat diet feeding induced downregulation of rictor gene expression. CONCLUSIONS: Here, we identify mTORC2 as a critical regulator of PVAT-directed protection of normal vascular tone. Modulation of mTORC2 activity in adipose tissue may be a potential therapeutic approach for inflammation-related vascular damage.


Subject(s)
Adipose Tissue/metabolism , Aorta, Thoracic/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Vasoconstriction , Vasodilation , 3T3-L1 Cells , Adipose Tissue/immunology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/immunology , Carrier Proteins/genetics , Chemokine CCL3/metabolism , Culture Media, Conditioned/metabolism , Cytokines/genetics , Diet, High-Fat , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inflammation/immunology , Inflammation/physiopathology , Interleukin-6/metabolism , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Real-Time Polymerase Chain Reaction , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Biochem Biophys Res Commun ; 429(1-2): 24-30, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23131564

ABSTRACT

The PIM serine/threonine kinases and the mTOR/AKT pathway integrate growth factor signaling and promote cell proliferation and survival. They both share phosphorylation targets and have overlapping functions, which can partially substitute for each other. In cancer cells PIM kinases have been reported to produce resistance to mTOR inhibition by rapamycin. Tumor growth depends highly on blood vessel infiltration into the malignant tissue and therefore on endothelial cell proliferation. We therefore investigated how the PIM1 kinase modulates growth inhibitory effects of rapamycin in mouse aortic endothelial cells (MAEC). We found that proliferation of MAEC lacking Pim1 was significantly more sensitive to rapamycin inhibition, compared to wildtype cells. Inhibition of mTOR and AKT in normal MAEC resulted in significantly elevated PIM1 protein levels in the cytosol and in the nucleus. We observed that truncation of the C-terminal part of Pim1 beyond Ser 276 resulted in almost exclusive nuclear localization of the protein. Re-expression of this Pim1 deletion mutant significantly increased the proliferation of Pim1(-/-) cells when compared to expression of the wildtype Pim1 cDNA. Finally, overexpression of the nuclear localization mutant and the wildtype Pim1 resulted in complete resistance to growth inhibition by rapamycin. Thus, mTOR inhibition-induced nuclear accumulation of PIM1 or expression of a nuclear C-terminal PIM1 truncation mutant is sufficient to increase endothelial cell proliferation, suggesting that nuclear localization of PIM1 is important for resistance of MAEC to rapamycin-mediated inhibition of proliferation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Nucleus/enzymology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-pim-1/metabolism , Sirolimus/pharmacology , Animals , Cells, Cultured , Endothelial Cells/drug effects , Mice , Proto-Oncogene Proteins c-pim-1/genetics , Sequence Deletion
6.
Cardiovasc Res ; 80(1): 106-13, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18566101

ABSTRACT

AIMS: Angiotensin converting enzyme (ACE) inhibition reduces heart disease and vascular stiffness in hypertension and leads to kinin accumulation. In this study, we analysed the role and importance of two kinin receptor subtypes in angiogenesis during ACE inhibition in an in vitro model of angiogenesis of the mouse heart. METHODS AND RESULTS: First, we analysed the angiogenic properties of bradykinin and enalapril on wild-type C57Bl/6 and B2 receptor(-/-) mouse heart under normoxia (21% O(2)) and hypoxia (1% O(2)) in vitro and the contribution of B1 and B2 kinin receptors to this effect. Bradykinin induced dose-dependent endothelial sprout formation in vitro in adult mouse heart only under hypoxia (1.7 fold, n = 6, P < 0.05). The B2 receptor mediated sprouting that was induced by bradykinin and vascular endothelial growth factor (VEGF(164); n = 6, P < 0.05), but did not mediate sprouting that was induced by growth factors bFGF or PDGF-BB. Enalapril induced sprouting through both the B1 and B2 kinin receptors, but it required the presence of the B2 receptor in both scenarios and was dependent on BK synthesis. B1-receptor agonists induced sprout formation via the B1 receptor (2.5 fold, n = 6, P < 0.05), but it required the presence of the B2 receptor for them to do so. Both B2-receptor and B1-receptor agonist-induced angiogenesis required nitric oxide biosynthesis. CONCLUSION: The kinin B2 receptor plays a crucial role in angiogenesis that is induced by different vasoactive molecules, namely bradykinin, ACE inhibitors, B1-stimulating kinin metabolites, and VEGF164 in an in vitro model of angiogenesis of mouse heart under hypoxia. Therapeutic treatment of hypertensive patients by using ACE inhibitors may potentially benefit the ischaemic heart through inducing B2-dependent heart neovascularization.


Subject(s)
Heart/physiology , Hypoxia/physiopathology , Neovascularization, Physiologic , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Bradykinin/physiology , Enalapril/pharmacology , Fibroblast Growth Factors/physiology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Nitric Oxide/biosynthesis , Receptor, Bradykinin B1/agonists , Receptor, Bradykinin B2/agonists , Vascular Endothelial Growth Factor A/physiology
7.
Hypertension ; 49(5): 1178-85, 2007 May.
Article in English | MEDLINE | ID: mdl-17339539

ABSTRACT

Angiotensin II is a vasoactive peptide that may affect vascularization of the ischemic heart via angiogenesis. In this study we aimed at studying the mechanisms underlying the angiogenic effects of angiotensin II under hypoxia in the mouse heart in vitro. Endothelial sprout formation from pieces of mouse hearts was assessed under normoxia (21% O(2)) and hypoxia (1% O(2)) during a 7-day period of in vitro culture. Only under hypoxia did angiotensin II dose-dependently induce endothelial sprout formation, peaking at 10(-7) mol/L of angiotensin II. Angiotensin II type 1 (AT(1)) receptor blockade by losartan did not affect angiotensin II-induced sprouting in wild-type mice. Conversely, the angiotensin II type 2 (AT(2)) receptor antagonist PD 123319 blocked this response. In hearts from AT(1)(-/-) mice, angiotensin II-elicited sprouting was preserved but blocked again by AT(2) receptor antagonism. In contrast, no angiotensin II-induced sprouting was found in preparations from hearts of AT(2)(-/-) mice. Angiotensin II-mediated angiogenesis was also abolished by a specific inhibitor of the B2 kinin receptor in both wild-type and AT(1)(-/-) mice. Furthermore, angiotensin II failed to induce endothelial sprout formation in hearts from B2(-/-) mice. Finally, NO inhibition completely blunted sprouting in hearts from wild-type mice, whereas NO donors could restore sprouting in AT(2)(-/-) and B2(-/-) hearts. This in vitro study suggests the obligatory role of hypoxia in the angiogenic effect of angiotensin II in the mouse heart via the AT(2) receptor through a mechanism that involves bradykinin, its B2 receptor, and NO as a downstream effector.


Subject(s)
Angiotensin II/pharmacology , Coronary Vessels/drug effects , Coronary Vessels/physiopathology , Hypoxia/physiopathology , Neovascularization, Physiologic , Receptor, Angiotensin, Type 2/metabolism , Receptor, Bradykinin B2/metabolism , Angiotensin II/administration & dosage , Animals , Dose-Response Relationship, Drug , Hypoxia/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/deficiency , Receptor, Bradykinin B2/deficiency
8.
Curr Opin Pharmacol ; 7(2): 151-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17276727

ABSTRACT

The microcirculation largely determines peripheral vascular resistance and substantially contributes to arterial hypertension. In both human arterial hypertension and animal models of hypertension, genetic, fetal and other mechanisms associated with hypertension can reduce the formation and number of microvessels (i.e. parallel-connected arterioles and capillaries). Impaired formation of microvessels (impaired angiogenesis) and microvascular rarefaction can, on the other hand, contribute to increased peripheral resistance and raise blood pressure. Interestingly, drugs targeting the renin-angiotensin-aldosterone system (i.e. angiotensin-converting enzyme inhibitors and AT(1) receptor blockers) induce angiogenesis in vivo in the majority of animal studies. Furthermore, recent clinical studies demonstrate that long-term antihypertensive treatment increases capillary density in the skin of hypertensive patients without diabetes. These effects of angiotensin-converting enzyme inhibitors and AT(1) receptor blockers can be mediated by activation of bradykinin pathways, resulting in the generation of vascular endothelial growth factor, nitric oxide and, consequently, angiogenesis. In conclusion, specific antihypertensive drugs can induce angiogenesis and reduce or even reverse microvascular rarefaction. This might improve target organ damage in, and slow the development of, hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Hypertension/drug therapy , Microcirculation/drug effects , Neovascularization, Physiologic/drug effects , Animals , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Blood Vessels/drug effects , Blood Vessels/physiopathology , Disease Models, Animal , Humans , Hypertension/physiopathology , Microcirculation/physiopathology , Neovascularization, Pathologic/drug therapy
9.
Circ Res ; 100(1): 79-87, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17110594

ABSTRACT

A central regulator of cell growth that has been implicated in responses to stress such as hypoxia is mTOR (mammalian Target Of Rapamycin). We have shown previously that mTOR is required for angiogenesis in vitro and endothelial cell proliferation in response to hypoxia. Here we have investigated mTOR-associated signaling components under hypoxia and their effects on cell proliferation in rat aortic endothelial cells (RAECs). Hypoxia (1% O(2)) rapidly (>30 minutes) and in a concentration-dependent manner promoted rapamycin-sensitive and sustained phosphorylation of mTOR-Ser2448 followed by nuclear translocation in RAECs. Similarly, hypoxia induced phosphorylation of the mTORC2 substrate Akt-Ser473 (3 to 6 hours at 1% O(2)) and a brief phosphorylation peak of the mTORC1 substrate S6 kinase-Thr389 (10 to 60 minutes). Phosphorylation of Akt was inhibited by mTOR knockdown and partially with rapamycin. mTOR knockdown, rapamycin, or Akt inhibition specifically and significantly inhibited proliferation of serum-starved RAECs under hypoxia (P<0.05; n> or =4). Similarly, hypoxia induced Akt-dependent and rapamycin-sensitive proliferation in mouse embryonic fibroblasts. This response was partially blunted by hypoxia-inducible factor-1alpha knockdown and not affected by TSC2 knockout. Finally, mTORC2 inhibition by rictor silencing, especially (P<0.001; n=7), and mTORC1 inhibition by raptor silencing, partially (P<0.05; n=7), inhibited hypoxia-induced RAEC proliferation. Thus, mTOR mediates an early response to hypoxia via mTORC1 followed by mTORC2, promoting endothelial proliferation mainly via Akt signaling. mTORC1 and especially mTORC2 might therefore play important roles in diseases associated with hypoxia and altered angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypoxia/metabolism , Hypoxia/pathology , Protein Kinases/metabolism , Transcription Factors/metabolism , Animals , Biological Transport , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Embryo, Mammalian , Fibroblasts/metabolism , Fibroblasts/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Osmolar Concentration , Phosphorylation , Protein Isoforms/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Serine , TOR Serine-Threonine Kinases , Threonine , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
10.
ALTEX ; 24 Spec No: 35-8, 2007.
Article in English | MEDLINE | ID: mdl-19835053

ABSTRACT

Controlled induction of the formation of new microvessels, i.e., therapeutic angiogenesis, may be used one day to treat patients that for example had suffered a myocardial infarction. Experimental models of angiogenesis in the heart in vivo substantially stress the animal. We therefore developed a model of angiogenesis of the heart in vitro, where mouse and rat heart pieces are stimulated under controlled conditions in a three dimensional matrix. Capillary-like sprouts emerging in these cultures represent early to midterm steps of angiogenesis and can be quantified to study potential angiogenic compounds and underlying mechanisms.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Capillaries/physiology , Cell Culture Techniques/methods , Coronary Circulation/physiology , Heart/physiology , Neovascularization, Physiologic , Angiotensin II/pharmacology , Animals , Becaplermin , Capillaries/cytology , Capillaries/drug effects , Coronary Circulation/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibrin/physiology , Fibroblast Growth Factor 2/pharmacology , Gels , Humans , Mice , Muscle Tonus/drug effects , Muscle Tonus/physiology , Myocardium/cytology , Myocardium/enzymology , Neovascularization, Physiologic/drug effects , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type II/deficiency , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Rats , Vasodilation/drug effects
11.
Am J Emerg Med ; 23(4): 474-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16032614

ABSTRACT

PURPOSE: It is controversial whether arterial hypertension (AHT) can be diagnosed in the emergency department (ED). We sought to prospectively investigate the natural time course of blood pressure (BP) to define an optimal period for AHT screening in ED patients with an elevated initial BP. PROCEDURES: Patients with a BP greater than 160/100 mm Hg upon ED admission underwent repeated BP measurements every 5 minutes for 2 hours using an automated device. Arterial hypertension was confirmed using 12-hour ambulatory BP measurement or repeated office BP measurement according to the Joint National Committee VII guidelines by the primary care physician after discharge from the hospital. MAIN FINDINGS: Systolic BP decreased significantly during the first 10 to 20 minutes of ED stay in hypertensive and normotensive patients without further significant changes thereafter. Diastolic BP remained stable in both hypertensive and normotensive patients. Discrimination between hypertensive and normotensive patients was best between minutes 60 and 80 after ED admission. An average BP of 165/105 mm Hg or higher during this period strongly suggests AHT whereas a BP of less than 130/80 mm Hg excludes AHT with high sensitivity. CONCLUSIONS: Screening for AHT in the ED is possible with high specificity and sensitivity. Blood pressure measurements between minutes 60 and 80 after entry into the ED yield the highest diagnostic value.


Subject(s)
Blood Pressure Determination/methods , Emergency Medicine/methods , Hypertension/diagnosis , Female , Humans , Male , Middle Aged , Prospective Studies , Reference Values , Sensitivity and Specificity
12.
Exp Cell Res ; 300(2): 272-82, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15474993

ABSTRACT

Neovascularization in the heart is usually investigated with models of angiogenesis in vivo. Here we present a simple model that allows investigating heart angiogenesis in mice and rats in vitro. Small pieces of left ventricular myocardium were cultured in three-dimensional fibrin gels for 10 days. A single mouse heart allowed assessing 24 conditions, each tested in octuplicates. Rat recombinant VEGF164, human recombinant bFGF, and human recombinant PDGF-BB were used under normoxia (21% O2) and hypoxia (3% O2), and outgrowth of endothelial sprouts from heart pieces was quantified. In 4-week-old OF1 mice, endothelial sprouts formed spontaneously. In contrast, in 12-week-old adult mice, virtually no sprouts formed under normoxia. Under hypoxia, sprout formation increased substantially. Different growth factors induced formation of distinct patterns of sprouts and unorganized single cells. Sprouts were composed of endothelial cells with smooth muscle cells or pericytes interacting with them, as assessed by immunohistochemistry. Taken together, our model is suited for investigation of angiogenesis of the heart in vitro. It may allow performing extensive series of experiments in vitro including rapid screening of pharmacological compounds and assessment of mechanisms of heart angiogenesis in transgenic animals in an easy straightforward manner.


Subject(s)
Biological Assay , Coronary Vessels/physiology , Heart/physiology , Neovascularization, Physiologic/physiology , Animals , Coronary Vessels/drug effects , Growth Substances/pharmacology , Heart/drug effects , Hypoxia/metabolism , Mice , Myocardium/cytology , Myocardium/metabolism , Neovascularization, Physiologic/drug effects , Rats , Time Factors
13.
FASEB J ; 16(8): 771-80, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12039858

ABSTRACT

Angiogenesis and vascular cell proliferation are pivotal in physiological and pathological processes including atherogenesis, restenosis, wound healing, and cancer development. Here we show that mammalian target of rapamycin (mTOR) signaling plays a key role in hypoxia-triggered smooth muscle and endothelial proliferation and angiogenesis in vitro. Hypoxia significantly increased DNA synthesis and proliferative responses to platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) in rat and human smooth muscle and endothelial cells. In an in vitro 3-dimensional model of angiogenesis, hypoxia increased PDGF- and FGF-stimulated sprout formation from rat and mouse aortas. Hypoxia did not modulate PDGF receptor mRNA, protein, or phosphorylation. PI3K activity was essential for cell proliferation under normoxic and hypoxic conditions. Activities of PI3K-downstream target PKB under hypoxia and normoxia were comparable. However, mTOR inhibition by rapamycin specifically abrogated hypoxia-mediated amplification of proliferation and angiogenesis, but was without effect on proliferation under normoxia. Accordingly, hypoxia-mediated amplification of proliferation was further augmented in mTOR-overexpressing endothelial cells. Thus, signaling via mTOR may represent a novel mechanism whereby hypoxia augments mitogen-stimulated vascular cell proliferation and angiogenesis.


Subject(s)
Cell Hypoxia/physiology , Muscle, Smooth, Vascular/blood supply , Neovascularization, Physiologic/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , 3T3 Cells , Animals , Cell Division/drug effects , Cells, Cultured , Chromones/pharmacology , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/pharmacology , Mice , Models, Biological , Morpholines/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Protein Kinases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Platelet-Derived Growth Factor/drug effects , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
14.
Blood Press ; 11(2): 116-24, 2002.
Article in English | MEDLINE | ID: mdl-12035872

ABSTRACT

Arterial hypertension (AH) is characterized by reduced nitric oxide (NO) biosynthesis, vasoconstriction, and reduced microvascular density. In this study we asked whether AH also reduces the number of microvessels by impairing angiogenesis. AH was induced in Dahl salt-sensitive rats (DSS) with a salt diet and in Wistar-Kyoto rats by inhibiting NO formation with Nomega-nitro-L-arginine (NNA). Three weeks after induction of AH, two wound chambers containing collagen I (Vitrogen) were sutured into the mesenteric cavity of each animal. After additional 14 days, wound chamber neovascularization and the extent of vascularized connective tissue ingrowth were quantified. In NNA-induced AH, the number of newly formed vessels and the ingrowth of vascularized connective tissue into the wound chamber decreased as compared to controls. However, the number of newly formed vessels and the ingrowth of vascularized connective tissue did not change with increasing blood pressure in salt-fed DSS rats as compared to those fed a normal diet. Inhibition of NO biosynthesis, but not necessarily elevating blood pressure, reduces angiogenesis. Microvascular rarefaction in AH may be partially due to reduced angiogenesis because of impaired NO biosynthesis.


Subject(s)
Hypertension/pathology , Hypertension/physiopathology , Neovascularization, Pathologic , Nitric Oxide/biosynthesis , Animals , Blood Pressure/physiology , Disease Models, Animal , Nitroarginine/pharmacology , Rats , Rats, Inbred Dahl , Rats, Inbred WKY , Sodium Chloride/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...