Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Cardiol ; 2012 Feb 13.
Article in English | MEDLINE | ID: mdl-22336256

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

2.
Circ Cardiovasc Genet ; 4(4): 349-58, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21642240

ABSTRACT

BACKGROUND: Dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) are classic forms of systolic and diastolic heart failure, respectively. Mutations in genes encoding sarcomere and cytoskeletal proteins are major causes of HCM and DCM. MURC, encoding muscle-restricted coiled-coil, a Z-line protein, regulates cardiac function in mice. We investigated potential causal role of MURC in human cardiomyopathies. METHODS AND RESULTS: We sequenced MURC in 1199 individuals, including 383 probands with DCM, 307 with HCM, and 509 healthy control subjects. We found 6 heterozygous DCM-specific missense variants (p.N128K, p.R140W, p.L153P, p.S307T, p.P324L, and p.S364L) in 8 unrelated probands. Variants p.N128K and p.S307T segregated with inheritance of DCM in small families (χ(2)=8.5, P=0.003). Variants p.N128K, p.R140W, p.L153P, and p.S364L were considered probably or possibly damaging. Variant p.P324L recurred in 3 independent probands, including 1 proband with a TPM1 mutation (p.M245T). A deletion variant (p.L232-R238del) was present in 3 unrelated HCM probands, but it did not segregate with HCM in a family who also had a MYH7 mutation (p.L907V). The phenotype in mutation carriers was notable for progressive heart failure leading to heart transplantation in 4 patients, conduction defects, and atrial arrhythmias. Expression of mutant MURC proteins in neonatal rat cardiac myocytes transduced with recombinant adenoviruses was associated with reduced RhoA activity, lower mRNA levels of hypertrophic markers and smaller myocyte size as compared with wild-type MURC. CONCLUSIONS: MURC mutations impart loss-of-function effects on MURC functions and probably are causal variants in human DCM. The causal role of a deletion mutation in HCM is uncertain.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Proteins/physiology , Mutation , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Hypertrophic/genetics , Case-Control Studies , DNA Mutational Analysis , Humans , Muscle Cells/metabolism , Muscle Cells/pathology , Muscle Proteins/genetics , Mutant Proteins/genetics , Rats , Transfection , Vesicular Transport Proteins
3.
Circ Res ; 105(11): 1149-58, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19834007

ABSTRACT

RATIONALE: It has been reported that interleukin (IL)-1 is associated with pathological cardiac remodeling and LV dilatation, whereas IL-1beta has also been shown to induce cardiomyocyte hypertrophy. Thus, the role of IL-1 in the heart remains to be determined. OBJECTIVE: We studied the role of hypertrophy signal-mediated IL-1beta/insulin-like growth factor (IGF)-1 production in regulating the progression from compensative pressure-mediated hypertrophy to heart failure. METHODS AND RESULTS: Pressure overload was performed by aortic banding in IL-1beta-deficient mice. Primarily cultured cardiac fibroblasts (CFs) and cardiac myocytes (CMs) were exposed to cyclic stretch. Heart weight, myocyte size, and left ventricular ejection fraction were significantly lower in IL-1beta-deficient mice (20%, 23% and 27%, respectively) than in the wild type 30 days after aortic banding, whereas interstitial fibrosis was markedly augmented. DNA microarray analysis revealed that IGF-1 mRNA level was markedly (approximately 50%) decreased in the IL-1beta-deficient hypertrophied heart. Stretch of CFs, rather than CMs, abundantly induced the generation of IL-1beta and IGF-1, whereas such IGF-1 induction was markedly decreased in IL-1beta-deficient CFs. IL-1beta released by stretch is at a low level unable to induce IL-6 but sufficient to stimulate IGF-1 production. Promoter analysis showed that stretch-mediated IL-1beta activates JAK/STAT to transcriptionally regulate the IGF-1 gene. IL-1beta deficiency markedly increased c-Jun N-terminal kinase (JNK) and caspase-3 activities and enhanced myocyte apoptosis and fibrosis, whereas replacement of IGF-1 or JNK inhibitor restored them. CONCLUSIONS: We demonstrate for the first time that pressure-mediated hypertrophy and mechanical stretch generates a subinflammatory low level of IL-1beta, which constitutively causes IGF-1 production to maintain adaptable compensation hypertrophy and inhibit interstitial fibrosis.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Insulin-Like Growth Factor I/metabolism , Interleukin-1beta/metabolism , Animals , Apoptosis/physiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypertrophy, Left Ventricular/pathology , Interleukin-1beta/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Janus Kinase 2/metabolism , Mice , Mice, Mutant Strains , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Interleukin-1/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Stress, Mechanical , Ventricular Pressure/physiology
4.
Proc Natl Acad Sci U S A ; 106(40): 17169-74, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19805158

ABSTRACT

An analysis of the clonality of cardiac progenitor cells (CPCs) and myocyte turnover in vivo requires genetic tagging of the undifferentiated cells so that the clonal marker of individual mother cells is traced in the specialized progeny. CPC niches in the atria and apex of the mouse heart were infected with a lentivirus carrying EGFP, and the destiny of the tagged cells was determined 1-5 months later. A common integration site was identified in isolated CPCs, cardiomyocytes, endothelial cells (ECs), and fibroblasts, documenting CPC self-renewal and multipotentiality and the clonal origin of the differentiated cell populations. Subsequently, the degree of EGFP-lentiviral infection of CPCs was evaluated 2-4 days after injection, and the number of myocytes expressing the reporter gene was measured 6 months later. A BrdU pulse-chasing protocol was also introduced as an additional assay for the analysis of myocyte turnover. Over a period of 6 months, each EGFP-positive CPC divided approximately eight times generating 230 cardiomyocytes; this value was consistent with the number of newly formed cells labeled by BrdU. To determine whether, human CPCs (hCPCs) are self-renewing and multipotent, these cells were transduced with the EGFP-lentivirus and injected after acute myocardial infarction in immunosuppressed rats. hCPCs, myocytes, ECs, and fibroblasts collected from the regenerated myocardium showed common viral integration sites in the human genome. Thus, our results indicate that the adult heart contains a pool of resident stem cells that regulate cardiac homeostasis and repair.


Subject(s)
Cell Differentiation , Cell Proliferation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Lineage , Clone Cells/cytology , Clone Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Lentivirus/genetics , Mice , Molecular Sequence Data , Myocardium/cytology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Rats , Rats, Inbred F344 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Proc Natl Acad Sci U S A ; 105(40): 15529-34, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18832173

ABSTRACT

The Notch receptor mediates cell fate decision in multiple organs. In the current work we tested the hypothesis that Nkx2.5 is a target gene of Notch1 and raised the possibility that Notch1 regulates myocyte commitment in the adult heart. Cardiac progenitor cells (CPCs) in the niches express Notch1 receptor, and the supporting cells exhibit the Notch ligand Jagged1. The nuclear translocation of Notch1 intracellular domain (N1ICD) up-regulates Nkx2.5 in CPCs and promotes the formation of cycling myocytes in vitro. N1ICD and RBP-Jk form a protein complex, which in turn binds to the Nkx2.5 promoter initiating transcription and myocyte differentiation. In contrast, transcription factors of vascular cells are down-regulated by Jagged1 activation of the Notch1 pathway. Importantly, inhibition of Notch1 in infarcted mice impairs the commitment of resident CPCs to the myocyte lineage opposing cardiomyogenesis. These observations indicate that Notch1 favors the early specification of CPCs to the myocyte phenotype but maintains the newly formed cells in a highly proliferative state. Dividing Nkx2.5-positive myocytes correspond to transit amplifying cells, which condition the replicative capacity of the heart. In conclusion, Notch1 may have critical implications in the control of heart homeostasis and its adaptation to pathologic states.


Subject(s)
Myocytes, Cardiac/cytology , Receptor, Notch1/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
6.
Mol Med ; 14(7-8): 436-42, 2008.
Article in English | MEDLINE | ID: mdl-18475309

ABSTRACT

Macrophages play a major role in the development of vascular lesions in atherogenesis. The cells express FcgammaRIIIa (CD16) identical to that in NK cells, but with a cell type-specific glycosylation, and these soluble forms (sFcgammaRIIIa) are present in plasma. We measured sFcgammaRIIIa(Mphi) derived from macrophages in plasma from subjects undergoing an annual medical checkup. The levels of sFcgammaRIIIa(Mphi) increased with age, and correlated positively with body mass index, blood pressure, LDL cholesterol to HDL cholesterol ratio, triglycerides, hemoglobin A1c, and creatinine, but negatively with HDL-cholesterol levels. The sFcgammaRIIIa(Mphi) levels were related to the number of risk factors for atherosclerosis: such as aging, current smoking, diabetes, hypertension, hyper-LDL-cholesterolemia, hypo-HDL-cholesterolemia, and family history of atherosclerotic diseases. In addition, the sFcgammaRIIIa(Mphi) levels were correlated with carotid maximum intima-media thickness (IMT). These findings indicate the macrophages are activated during the incipient stage of atherosclerosis, and suggest sFcgammaRIIIa(Mphi) may be used as a predictive marker for atherosclerosis.


Subject(s)
Carotid Arteries/anatomy & histology , Receptors, IgG/blood , Tunica Intima/anatomy & histology , Adult , Atherosclerosis/blood , Atherosclerosis/diagnosis , Female , Humans , Male , Middle Aged , Models, Biological , Physical Examination , Prognosis , Solubility
7.
Circulation ; 116(9): 1041-51, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17698736

ABSTRACT

BACKGROUND: The involvement of Ca2+-dependent tyrosine kinase PYK2 in the Akt/endothelial NO synthase pathway remains to be determined. METHODS AND RESULTS: Blood flow recovery and neovessel formation after hind-limb ischemia were impaired in PYK2-/- mice with reduced mobilization of endothelial progenitors. Vascular endothelial growth factor (VEGF)-mediated cytoplasmic Ca2+ mobilization and Ca2+-independent Akt activation were markedly decreased in the PYK2-deficient aortic endothelial cells, whereas the Ca2+-independent AMP-activated protein kinase/protein kinase-A pathway that phosphorylates endothelial NO synthase was not impaired. Acetylcholine-mediated aortic vasorelaxation and cGMP production were significantly decreased. Vascular endothelial growth factor-dependent migration, tube formation, and actin cytoskeletal reorganization associated with Rac1 activation were inhibited in PYK2-deficient endothelial cells. PI3-kinase is associated with vascular endothelial growth factor-induced PYK2/Src complex, and inhibition of Src blocked Akt activation. The vascular endothelial growth factor-mediated Src association with PLCgamma1 and phosphorylation of 783Tyr-PLCgamma1 also were abolished by PYK2 deficiency. CONCLUSION: These findings demonstrate that PYK2 is closely involved in receptor- or ischemia-activated signaling events via Src/PLCgamma1 and Src/PI3-kinase/Akt pathways, leading to endothelial NO synthase phosphorylation, and thus modulates endothelial NO synthase-mediated vasoactive function and angiogenic response.


Subject(s)
Focal Adhesion Kinase 2/physiology , Heart/physiology , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/metabolism , Oncogene Protein v-akt/physiology , Analysis of Variance , Animals , Calcium/physiology , Enzyme Activation , Focal Adhesion Kinase 2/deficiency , Hindlimb/blood supply , Ischemia/physiopathology , Mice , Mice, Knockout , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor A/physiology , Vasodilation
8.
J Mol Cell Cardiol ; 36(4): 469-80, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15081307

ABSTRACT

We have reported that interleukin-1 beta (IL-1 beta) upregulates cardiac expression of vascular endothelial growth factor (VEGF) and VEGF receptor-2 (VEGFR-2), raising the possibility that IL-1 beta plays an important role in VEGF-mediated neovascularization. In this study, we examined the cellular mechanism for ischemia-induced neovascularization using IL-1 beta knock-out (-/-) mice. Recovery of blood perfusion in ischemic hindlimb in IL-1 beta-/- mice was markedly (43% decrease) impaired as compared with the wild-type mice. CD31(+) vessel numbers and Ki-67(+) neo-capillaries were significantly (P < 0.01) decreased 44% and 68%, respectively. IL-1 beta expression was localized in the capillary vessels in ischemic limb muscles. Ischemia-induced expressions of hypoxia-inducible factor 1 alpha (HIF-1 alpha), VEGF, its receptor VEGFR-2 and vascular cell adhesion molecule-1 (VCAM-1) were markedly inhibited in the IL-1 beta-/- mice. Hindlimb ischemia-induced an increase (1.22% out of total nuclear cell) in CD34(-)/B220(-)/CD3(-)/Flk-1(+) hematopoietic stem cell population in peripheral blood in the wild-type mice, whereas in the IL-1 beta-/- mice such increase was only 0.09%. Injection of IL-1 beta protein into the wild-type mice markedly increased the ratio of the CD34(-)/B220(-)/CD3(-)/Flk-1(+) cell population (from 0.03% to 0.7%) in the peripheral blood associated with an increase in the number of endothelial cells. Such IL-1 beta-mediated increases in cell numbers were blocked by co-injection of anti-VEGF antibody. CD34(-)/B220(-)CD3(-)Flk-1(+) cells trans-differentiated into eNOS- and CD31-expressing endothelial cells in vivo and in vitro. This study demonstrates that IL-1 beta plays a key role in ischemia-induced neovascularization by mobilizing CD34(-)/B220(-)CD3(-)Flk-1(+) endothelial precursor cells in a VEGF-dependent manner as well as by upregulating expressions of VEGF, VEGFR-2 and adhesion molecules on endothelial cells.


Subject(s)
Interleukin-1/genetics , Interleukin-1/physiology , Neovascularization, Pathologic , Animals , Antigens, CD34/biosynthesis , Blotting, Western , CD3 Complex/biosynthesis , Calibration , Cell Differentiation , Cell Separation , Endothelium, Vascular/metabolism , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Interleukin-1/metabolism , Ischemia , Ki-67 Antigen/biosynthesis , Laser-Doppler Flowmetry , Leukocyte Common Antigens/biosynthesis , Male , Mice , Mice, Knockout , Mice, Transgenic , Perfusion , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factors/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Circ Res ; 93(10): 980-9, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14525810

ABSTRACT

Peripheral blood (PB)-derived CD14+ monocytes were shown to transdifferentiate into endothelial cell (EC) lineage cells and contribute to neovascularization. We investigated whether bone marrow (BM)- or PB-derived CD34-/CD14+ cells are involved in reendothelialization after carotid balloon injury. Although neither hematopoietic nor mesenchymal stem cells were included in human BM-derived CD34-/CD14+ monocyte lineage cells (BM-MLCs), they expressed EC-specific markers (Tie2, CD31, VE-cadherin, and endoglin) to an extent identical to mature ECs. When BM-MLCs were cultured with vascular endothelial growth factors, hematopoietic markers were drastically decreased and new EC-specific markers (Flk and CD34) were induced. BM-MLCs were intra-arterially transplanted into balloon-injured arteries of athymic nude rats. When BM-MLCs were activated by monocyte chemoattractant protein-1 (MCP-1) in vivo or in vitro, they adhered onto injured endothelium, differentiated into EC-like cells by losing hematopoietic markers, and inhibited neointimal hyperplasia. Ability to prevent neointimal hyperplasia was more efficient than that of BM-derived CD34+ cells. MCP-dependent adhesion was not observed in PB-derived CD34-/CD14+ monocytes. Regenerated endothelium exhibited a cobblestone appearance, blocked extravasation of dye, and induced NO-dependent vasorelaxation. Basal adhesive activities on HUVECs under laminar flow and beta1-integrin expression (basal and active forms) were significantly increased in BM-MLCs compared with PB-derived monocytes. MCP-1 markedly enhanced adhesive activity of BM-MLCs (2.8-fold) on HUVECs by activating beta1-integrin conformation. Thus, BM-MLCs can function as EC progenitors that are more potent than CD34+ cells and acquire the ability to adhere on injured endothelium in a MCP-1-dependent manner, leading to reendothelialization associated with inhibition of intimal hyperplasia. This will open a novel window to MCP-1-mediated biological actions and vascular regeneration strategies by cell therapy.


Subject(s)
Bone Marrow Cells/cytology , Chemokine CCL2/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Monocytes/physiology , Stem Cells/physiology , Angioplasty, Balloon/adverse effects , Animals , Antigens, Differentiation/biosynthesis , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion/physiology , Cell Differentiation , Cell Lineage , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/pharmacology , Endothelium, Vascular/immunology , Endothelium, Vascular/injuries , Gene Transfer Techniques , Humans , Monocytes/cytology , Monocytes/immunology , Rats , Rats, Nude , Stem Cells/cytology
10.
Hypertension ; 41(1): 99-107, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511537

ABSTRACT

We have previously demonstrated that stimulation of the angiotensin (Ang) II type 2 receptor in vascular smooth muscle cells caused bradykinin production by activating kininogenase in transgenic mice. The aim of this study was to determine whether overexpression of AT2 receptors in cardiomyocytes attenuates Ang II-induced cardiomyocyte hypertrophy or interstitial fibrosis through a kinin/nitric oxide (NO)-dependent mechanism in mice. Ang II (1.4 mg/kg per day) or vehicle was subcutaneously infused into transgenic mice and wild-type mice for 14 days. The amount of cardiac AT2 receptor relative to AT1 receptor in transgenic mice was 22% to 37%. Ang II caused similar elevations in systolic blood pressure (by approximately 45 mm Hg) in transgenic mice and wild-type mice. Myocyte hypertrophy assessed by an increase in myocyte cross-sectional area, left ventricular mass, and atrial natriuretic peptide mRNA levels were similar in transgenic and wild-type mice. Ang II induced prominent perivascular fibrosis of the intramuscular coronary arteries, the extent of which was significantly less in transgenic mice than in wild-type mice. Inhibition of perivascular fibrosis in transgenic mice was abolished by cotreatment with HOE140, a bradykinin B2 receptor antagonist, or L-NAME, an inhibitor of NO synthase. Cardiac kininogenase activity was markedly increased (approximately 2.6-fold, P<0.001) after Ang II infusion in transgenic mice but not in wild-type mice. Immunohistochemistry indicated that both bradykinin B2 receptors and endothelial NO synthase were expressed in the vascular endothelium, whereas only B2 receptors were present in fibroblasts. These results suggest that stimulation of AT2 receptors present in cardiomyocytes attenuates perivascular fibrosis by a kinin/NO-dependent mechanism. However, the effect on the development of cardiomyocyte hypertrophy was not detected in this experimental setting.


Subject(s)
Bradykinin/metabolism , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Nitric Oxide/metabolism , Receptors, Angiotensin/physiology , Angiotensin II/pharmacology , Animals , Coronary Vessels/pathology , Extracellular Space , Fibrosis , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/metabolism , Kallikreins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase/analysis , RNA, Messenger/biosynthesis , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptor, Bradykinin B2 , Receptors, Angiotensin/analysis , Receptors, Angiotensin/genetics , Receptors, Bradykinin/analysis , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
11.
Hypertension ; 41(1): 156-62, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511546

ABSTRACT

It remains undetermined whether continuous endothelial nitric oxide (NO) overexpression exerts angiogenic action. We surgically induced hindlimb ischemia in transgenic mice overexpressing endothelial NO synthase in the endothelium (eNOS-Tg) and studied neocapillary formation, ischemia-induced vascular endothelial growth factor (VEGF) expression, cGMP accumulation, and Akt/PKB signaling. Laser Doppler imaging revealed a markedly increased recovery of blood perfusion in ischemic limbs of eNOS-Tg mice (44% increase) compared with that in wild-type mice. Angiography showed a marked increase in basal and ischemia-induced collateral vessel formation in eNOS-Tg mice. Basal capillary densities and tissue cGMP levels were increased in eNOS-Tg mice (1.8-fold and 1.6-fold versus wild-type mice, respectively). Ischemia-induced neocapillary formation and cGMP accumulation were markedly increased in eNOS-Tg mice (3.6-fold and 4.1-fold versus preischemia levels, respectively), whereas those in wild-type mice were much less (1.8-fold and 1.5-fold, respectively). Basal and time-dependent VEGF expression in ischemic muscles did not differ between eNOS-Tg and wild-type mice. Basal and VEGF-mediated Akt phosphorylation in aortas was similar between eNOS-Tg and wild-type mice. Aortic basal eNOS expression was increased 3.3-fold, and VEGF-mediated eNOS phosphorylation was markedly induced in aortas of eNOS-Tg compared with preischemia levels (4.2-fold), whereas much smaller changes were observed in wild-type mice (1.8-fold increase). Our study demonstrates that overexpression of eNOS protein causes a marked increase in neocapillary formation in response to tissue ischemia without affecting ischemia-induced VEGF expression or VEGF-mediated Akt phosphorylation.


Subject(s)
Ischemia/blood , Neovascularization, Physiologic , Nitric Oxide Synthase/genetics , Protein Serine-Threonine Kinases , Angiography , Animals , Blood Circulation , Capillaries/growth & development , Collateral Circulation , Cyclic GMP/analysis , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Hindlimb/blood supply , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Ischemia/enzymology , Ischemia/metabolism , Laser-Doppler Flowmetry , Lymphokines/genetics , Lymphokines/pharmacology , Male , Mice , Mice, Transgenic , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Arterioscler Thromb Vasc Biol ; 22(11): 1804-10, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12426208

ABSTRACT

OBJECTIVE: This study was performed to evaluate the angiogenic effect of implantation of peripheral blood mononuclear cells (PB-MNCs) compared with bone marrow mononuclear cells (BM-MNCs) into ischemic hibernating myocardium. METHODS AND RESULTS: A NOGA electromechanical system was used to map the hibernating region and to inject cells. PB-MNCs and BM-MNCs contained similar levels of vascular endothelial growth factor and basic fibroblast growth factor, whereas contents of angiogenic cytokines (interleukin-1beta and tumor necrosis factor-alpha) were larger in PB-MNCs. Numbers of endothelial progenitors were approximately 500-fold higher in BM-MNCs. In BM-MNC-implanted myocardia of pigs, an increase in systolic function (ejection fraction from 33% to 52%) and regional blood flow (2.1-fold) and a reduction of the ischemic area (from 29% to 8%) were observed. PB-MNC implantation reduced the ischemic area (from 31% to 17%), the extent of which was less than that seen with BM-MNCs. In saline-implanted myocardium, the ischemic area expanded (from 28% to 38%), and systolic function deteriorated. Angiography revealed an increase in collateral vessel formation by PB-MNC or BM-MNC implantation. Capillary numbers were increased 2.6- and 1.7-fold by BM-MNC and PB-MNC implantation, respectively. BM-MNCs but not PB-MNCs were incorporated into neocapillaries. CONCLUSIONS: Catheter-based implantation of PB-MNCs can effectively improve collateral perfusion and regional function in hibernating ischemic myocardium by its ability to mainly supply angiogenic factors and cytokines.


Subject(s)
Coronary Circulation/physiology , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/transplantation , Myocardial Contraction/physiology , Myocardial Ischemia/therapy , Perfusion/methods , Angiogenesis Inducing Agents/metabolism , Animals , Bone Marrow Cells/chemistry , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Capillaries/chemistry , Capillaries/cytology , Cardiac Catheterization/methods , Cell Lineage , Coronary Angiography/methods , Coronary Vessels/chemistry , Coronary Vessels/cytology , Coronary Vessels/physiology , Electrophysiologic Techniques, Cardiac/methods , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Immunophenotyping/methods , Leukocytes, Mononuclear/chemistry , Neovascularization, Physiologic/physiology , Recovery of Function/physiology , Stem Cell Transplantation/methods , Stem Cells/chemistry , Stem Cells/metabolism , Stem Cells/physiology , Swine
13.
Circulation ; 106(15): 2019-25, 2002 Oct 08.
Article in English | MEDLINE | ID: mdl-12370229

ABSTRACT

BACKGROUND: Peripheral blood mononuclear cells (PBMNCs), platelets, and polymorphonuclear leukocytes (PMNs) contain various angiogenic factors and cytokines. METHODS AND RESULTS: Unilateral hindlimb ischemia was surgically induced in athymic nude rats, and fluorescence-labeled human blood cells (PBMNCs [10(7) cells]+platelets [10(9)] or PBMNCs [10(7)]+platelets [10(9)]+PMNs [10(7)]) were intramuscularly implanted into the ischemic limbs. Laser Doppler imaging revealed markedly increased blood perfusion in PBMNC+platelet-implanted limbs (44% increase, P<0.001) compared with control implantation of human umbilical vein vascular endothelial cells. The addition of PMNs to PBMNCs+platelets attenuated blood perfusion (27% decrease, P<0.01). Neocapillary densities were increased by implantation of PBMNCs+platelets or platelets alone (3.5-fold and 2.4-fold, respectively; P<0.001), whereas PMNs inhibited (32%, P<0.05) PBMNC+ platelet-mediated capillary formation. There was no incorporation of implanted PBMNCs into neocapillaries, whereas PBMNCs and platelets accumulated around arterioles after implantation. Cellular extract from PBMNCs+platelets, in which vascular endothelial growth factor (VEGF), basic fibroblast growth factor, platelet-derived growth factor-AB, and transforming growth factor-beta were detected, markedly stimulated tubule formation of human umbilical vein vascular endothelial cells. Anti-VEGF neutralizing antibody markedly inhibited tubule formation and in vivo vessel formation. Neutrophil elastase inhibitor blocked the antiangiogenic action of PMNs, whereas inhibitors of oxygen metabolites had no effect. CONCLUSIONS: This study demonstrated that implantation of PBMNCs and platelets into ischemic limbs effectively induces collateral vessel formation by supplying angiogenic factors (mainly VEGF) and cytokines, suggesting that this cell therapy is useful as a novel strategy for therapeutic angiogenesis.


Subject(s)
Ischemia/surgery , Leukocytes, Mononuclear/transplantation , Neovascularization, Physiologic , Platelet Transfusion , Angiogenesis Inducing Agents/physiology , Angiography , Animals , Blood Circulation , Blood Platelets/physiology , Blood Vessels/chemistry , Blood Vessels/growth & development , Bone Marrow Transplantation , Cell Movement , Endothelial Growth Factors/physiology , Endothelium, Vascular/growth & development , Extremities/blood supply , Factor VIII/analysis , Factor VIII/immunology , Humans , Immunohistochemistry , Ischemia/diagnostic imaging , Ischemia/physiopathology , Laser-Doppler Flowmetry , Leukocytes, Mononuclear/physiology , Lymphokines/physiology , Neutrophils/physiology , Neutrophils/transplantation , Rats , Rats, Nude , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Nihon Rinsho ; 60(10): 1911-5, 2002 Oct.
Article in Japanese | MEDLINE | ID: mdl-12397683

ABSTRACT

UNLABELLED: We tested angiogenic activities of angiotensin II(Ang II) in ischemic hindlimbs using AT1 receptor(AT1R)-knock out(KO), AT2R-KO, wild-type(WT) mice. METHODS AND RESULTS: Angiogenesis was evaluated three weeks after unilateral hindlimb ischemia by laser Doppler perfusion(LDP) and capillary density. The ischemia/normal LDP ratio was markedly(p < 0.001) decreased in AT1R-KO(54 +/- 5% recovery) and AngII infusion-AT1R-KO(43 +/- 3%) than in WT(71 +/- 6%). In contrast, ischemia/normal LDP ratio was significantly(p < 0.01) increased in AT2R-KO(82 +/- 5%) and AngII infusion-AT2R-KO(96 +/- 6%) than in WT(71 +/- 6%). AT1R-KO and AngII infusion -AT1R-KO mice displayed lower capillary densities than WT(15 +/- 3, 11 +/- 3 vs 24 +/- 3 per field; p < 0.001). CONCLUSION: Ischemia in skeletal muscle causes upregulation of AT1R and AT2R expression, which positively and negatively modulates VEGF expression. This VEGF regulation via AngII receptor subtypes is closely involved in postnatal angiogenesis in ischemic limbs.


Subject(s)
Angiotensin II/physiology , Neovascularization, Physiologic/physiology , Receptors, Angiotensin/physiology , Animals , Mice , Mice, Knockout , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2
15.
Lancet ; 360(9331): 427-35, 2002 Aug 10.
Article in English | MEDLINE | ID: mdl-12241713

ABSTRACT

BACKGROUND: Preclinical studies have established that implantation of bone marrow-mononuclear cells, including endothelial progenitor cells, into ischaemic limbs increases collateral vessel formation. We investigated efficacy and safety of autologous implantation of bone marrow-mononuclear cells in patients with ischaemic limbs because of peripheral arterial disease. METHODS: We first did a pilot study, in which 25 patients (group A) with unilateral ischaemia of the leg were injected with bone marrow-mononuclear cells into the gastrocnemius of the ischaemic limb and with saline into the less ischaemic limb. We then recruited 22 patients (group B) with bilateral leg ischaemia, who were randomly injected with bone marrow-mononuclear cells in one leg and peripheral blood-mononuclear cells in the other as a control. Primary outcomes were safety and feasibility of treatment, based on ankle-brachial index (ABI) and rest pain, and analysis was per protocol. FINDINGS: Two patients were excluded from group B after randomisation. At 4 weeks in group B patients, ABI was significantly improved in legs injected with bone marrow-mononuclear cells compared with those injected with peripheral blood-mononuclear cells (difference 0.09 [95% CI 0.06-0.11]; p<0.0001). Similar improvements were seen for transcutaneous oxygen pressure (13 [9-17]; p<0.0001), rest pain (-0.85 [-1.6 to -0.12]; p=0.025), and pain-free walking time (1.2 [0.7-1.7]; p=0.0001). These improvements were sustained at 24 weeks. Similar improvements were seen in group A patients. Two patients in group A died after myocardial infarction unrelated to treatment. INTERPRETATION: Autologous implantation of bone marrow-mononuclear cells could be safe and effective for achievement of therapeutic angiogenesis, because of the natural ability of marrow cells to supply endothelial progenitor cells and to secrete various angiogenic factors or cytokines.


Subject(s)
Bone Marrow Transplantation , Extremities/blood supply , Ischemia/therapy , Leukocytes, Mononuclear/transplantation , Neovascularization, Physiologic/physiology , Aged , Angiogenesis Inducing Agents/analysis , Feasibility Studies , Female , Foot/blood supply , Humans , Immunohistochemistry , Male , Pilot Projects , Transplantation, Autologous , Treatment Outcome
16.
Kidney Int ; 62(3): 799-808, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12164862

ABSTRACT

BACKGROUND: Gq-coupled receptors are known to transactivate epidermal growth factor receptor (EGFR) via the Ca2+ and PKC pathways to phosphorylate extracellular signal-regulated kinase (ERK). METHODS: We studied the involvement of EGFR in transforming growth factor-beta (TGF-beta)-mediated fibronectin (FN) expression using glomerular mesangial cells. RESULTS: TGF-beta up-regulated FN mRNA accumulation in a time- and dose-dependent manner, which was completely inhibited by phosphatidylcholine-phospholipase C (PC-PLC) inhibitor and PKC inhibitors (calphostin-C and staurosporin). The EGFR inhibitor AG1478 completely abolished TGF-beta-mediated FN expression. ERK inactivation by PD98059, and p38MAPK inhibitor SB203580 also showed significant inhibitory effects. Addition of neutralizing anti-heparin-binding EGF-like growth factor (HB-EGF) antibody, pretreatment with heparin and the metalloproteinase (MMP) inhibitor batimastat blocked FN expression. In mesangial cells stably transfected with a chimera containing HB-EGF and alkaline phosphatase (ALP) genes, ALP activity in incubation medium was rapidly increased by TGF-beta (2.1-fold at 0.5 min) and reached a 3.7-fold increase at two minutes, which was abolished by calphostin-C or batimastat. TGF-beta phosphorylated EGFR, ERK and p38MAPK in a PKC- and MMP-dependent manner. Smad2 phosphorylation by TGF-beta was not affected by AG1478, and HB-EGF did not activate Smad2. FN mRNA stability was not affected by TGF-beta. Cycloheximde did not interfere with TGF-beta-mediated FN expression. CONCLUSIONS: The present study demonstrated that HB-EGF processed and released via PC-PLC-PKC signaling is an intermediate molecule for TGF-beta-mediated EGFR transactivation, and subsequent activation of ERK and p38MAPK is involved in FN expression via transcriptional regulation without requiring new protein synthesis.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Fibronectins/genetics , Glomerular Mesangium/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Cells, Cultured , Fibronectins/metabolism , Glomerular Mesangium/cytology , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , RNA, Messenger/analysis , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Type C Phospholipases/metabolism , Up-Regulation , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...