301
|
Experimental study of the effects of marrow mesenchymal stem cells transfected with hypoxia-inducible factor-1alpha gene. J Biomed Biotechnol 2009; 2009:128627. [PMID: 19587827 PMCID: PMC2705776 DOI: 10.1155/2009/128627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 04/09/2009] [Accepted: 04/15/2009] [Indexed: 12/03/2022] Open
Abstract
Objective. To construct the eukaryotic expression
vector hypoxia-inducible factor
1α-pcDNA3.1 and to investigate its transfective efficiency into
mesenchymal stem cells (MSCs) in vitro and the expression of
HIF-1α gene in MSCs.
Methods. mRNA of Wistar Rats' myocardial cells
was extracted, and cDNA was synthesized with Reverse Transcription
Kit, HIF-1α was
amplified by polymerase chain reaction (PCR), and constructed into
pcDNA3.1. Transfected
HIF-1α-pcDNA3.1 into MSCs by liposome mediated method. The expression
of HIF-1α in the
cells was detected by Western Blot Analysis and ELISA.
Results. Eukaryotic expression vector
HIF-1α-pcDNA3.1 was constructed successfully. Analyzed by flow
cytometer, The MSCs' surfaces mark were CD44+, SH3(CD73)+,
CD34−, CD45− and the CD44+ cells and SH3(CD73)+ cells were
94.7% and 97.3%, respectively, showing the high purity of
the cultured MSCs. After inducing, the cultured MSCs can
differentiate into osteoblasts and adipocytes successfully. In
HIF-1α gene
transfected MSCs, the expression of
HIF-1α mRNA and
HIF-1α protein were
both increased obviously. Conclusion.
HIF-1α was cloned
successfully.
HIF-1α-pcDNA3.1 can be transfected into MSCs by liposome-mediated
method effectively and which resulting stable expression of
HIF-1α in transfected
MSCs.
Collapse
|
302
|
Guo Q, Carrero JJ, Yu X, Barany P, Qureshi AR, Eriksson M, Anderstam B, Chmielewski M, Heimburger O, Stenvinkel P, Lindholm B, Axelsson J. Associations of VEGF and its receptors sVEGFR-1 and -2 with cardiovascular disease and survival in prevalent haemodialysis patients. Nephrol Dial Transplant 2009; 24:3468-73. [DOI: 10.1093/ndt/gfp315] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
303
|
van der Laan AM, Piek JJ, van Royen N. Targeting angiogenesis to restore the microcirculation after reperfused MI. Nat Rev Cardiol 2009; 6:515-23. [DOI: 10.1038/nrcardio.2009.103] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
304
|
Chen SY, Wang F, Yan XY, Zhou Q, Ling Q, Ling JX, Rong YZ, Li YG. Autologous transplantation of EPCs encoding FGF1 gene promotes neovascularization in a porcine model of chronic myocardial ischemia. Int J Cardiol 2009; 135:223-32. [DOI: 10.1016/j.ijcard.2008.12.193] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Revised: 11/19/2008] [Accepted: 12/13/2008] [Indexed: 01/08/2023]
|
305
|
Enhanced mobilization of CD34+ progenitor cells expressing cell adhesion molecules in patients with STEMI. Clin Res Cardiol 2009; 98:477-86. [DOI: 10.1007/s00392-009-0021-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 04/16/2009] [Indexed: 10/20/2022]
|
306
|
Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev 2009; 89:481-534. [PMID: 19342613 DOI: 10.1152/physrev.00042.2007] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide production in response to flow-dependent shear forces applied on the surface of endothelial cells is a fundamental mechanism of regulation of vascular tone, peripheral resistance, and tissue perfusion. This implicates the concerted action of multiple upstream "mechanosensing" molecules reversibly assembled in signalosomes recruiting endothelial nitric oxide synthase (eNOS) in specific subcellular locales, e.g., plasmalemmal caveolae. Subsequent short- and long-term increases in activity and expression of eNOS translate this mechanical stimulus into enhanced NO production and bioactivity through a complex transcriptional and posttranslational regulation of the enzyme, including by shear-stress responsive transcription factors, oxidant stress-dependent regulation of transcript stability, eNOS regulatory phosphorylations, and protein-protein interactions. Notably, eNOS expressed in cardiac myocytes is amenable to a similar regulation in response to stretching of cardiac muscle cells and in part mediates the length-dependent increase in cardiac contraction force. In addition to short-term regulation of contractile tone, eNOS mediates key aspects of cardiac and vascular remodeling, e.g., by orchestrating the mobilization, recruitment, migration, and differentiation of cardiac and vascular progenitor cells, in part by regulating the stabilization and transcriptional activity of hypoxia inducible factor in normoxia and hypoxia. The continuum of the influence of eNOS in cardiovascular biology explains its growing implication in mechanosensitive aspects of integrated physiology, such as the control of blood pressure variability or the modulation of cardiac remodeling in situations of hemodynamic overload.
Collapse
Affiliation(s)
- J-L Balligand
- Unit of Pharmacology and Therapeutics, Université catholique de Louvain, Brussels, Belgium.
| | | | | |
Collapse
|
307
|
Abstract
Molecular imaging represents a targeted approach to noninvasively assess biologic (both physiologic and pathologic) processes in vivo. Ideally the goal of molecular imaging is not just to provide diagnostic and prognostic information based on identification of the molecular events associated with a pathologic process but rather to guide individually tailored pharmacologic, cell-based, or genetic therapeutic regimens. This article reviews the recent advances in myocardial molecular imaging in the context of the cardiovascular processes of angiogenesis, apoptosis, inflammation, and ventricular remodeling. The focus is on radiotracer-based single photon emission computed tomography and positron emission tomography molecular imaging approaches.
Collapse
Affiliation(s)
- Alan R Morrison
- Yale University School of Medicine, Section of Cardiovascular Medicine, New Haven, CT 06520-8017, USA
| | | |
Collapse
|
308
|
Schwartz AL, Ciechanover A. Targeting proteins for destruction by the ubiquitin system: implications for human pathobiology. Annu Rev Pharmacol Toxicol 2009; 49:73-96. [PMID: 18834306 DOI: 10.1146/annurev.pharmtox.051208.165340] [Citation(s) in RCA: 362] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cellular proteins are in a dynamic state maintained by synthesis and degradation. The ubiquitin proteolytic pathway is responsible for the degradation of the bulk of cellular proteins including short-lived, regulatory, and misfolded/denatured proteins. Ubiquitin-mediated proteolysis involves covalent attachment of multiple ubiquitin molecules to the protein substrate and degradation of the targeted protein by the 26S proteasome. Recent understanding of the molecular mechanisms involved provides a framework to understand a wide variety of human pathophysiological states as well as therapeutic interventions. This review focuses on the response to hypoxia, inflammatory diseases, neurodegenerative diseases, and muscle-wasting disorders, as well as human papillomaviruses, cervical cancer and other malignancies.
Collapse
Affiliation(s)
- Alan L Schwartz
- Department of Pediatrics, Washington University School of Medicine and St. Louis Children's Hospital, St. Louis, Missouri 63110, USA.
| | | |
Collapse
|
309
|
Henderson KK, Danzi S, Paul JT, Leya G, Klein I, Samarel AM. Physiological replacement of T3 improves left ventricular function in an animal model of myocardial infarction-induced congestive heart failure. Circ Heart Fail 2009; 2:243-52. [PMID: 19808346 DOI: 10.1161/circheartfailure.108.810747] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Patients with congestive heart failure (CHF) often have low serum triiodothyronine (T(3)) concentrations. In a rodent model of myocardial infarction-induced CHF and low serum T(3), we hypothesized that replacing T(3) to euthyroid levels would improve left ventricular function without producing untoward signs of thyrotoxicosis. METHODS AND RESULTS Adult male Sprague-Dawley rats were subjected to left anterior descending coronary artery ligation (myocardial infarction). One week post-myocardial infarction, left ventricular fractional shortening was significantly reduced to 22+/-1% in CHF animals versus 38+/-1% for sham-operated controls (P<0.001). Serum T(3) concentration was also significantly reduced (80+/-3 versus 103+/-6 ng/dL; P<0.001), in CHF animals versus Shams. At 9 weeks post-myocardial infarction, systolic function (+dP/dt max) was significantly attenuated in CHF animals (4773+/-259 versus 6310+/-267 mmHg/s; P<0.001) as well as diastolic function measured by half time to relaxation (15.9+/-1.2 versus 11.1+/-0.3 ms; P<0.001). alpha-myosin heavy chain expression was also significantly reduced by 77% (P<0.001), and beta-myosin heavy chain expression was increased by 21%. Continuous T(3) replacement was initiated 1 week post-myocardial infarction with osmotic mini-pumps (6 microg/kg/d), which returned serum T(3) concentrations to levels similar to Sham controls while resting conscious heart rate, arterial blood pressure and the incidence of arrhythmias were not different. At 9 weeks, systolic function was significantly improved by T(3) replacement (6279+/-347 mmHg/s; P<0.05) and a trend toward improved diastolic function (12.3+/-0.6 ms) was noted. T(3) replacement in CHF animals also significantly increased alpha- and reduced beta-MHC expression, (P<0.05). CONCLUSIONS These data indicate that T(3) replacement to euthyroid levels improves systolic function and tends to improve diastolic function, potentially through changes in myocardial gene expression.
Collapse
MESH Headings
- Animals
- Blood Pressure
- Diastole
- Disease Models, Animal
- Gene Expression Regulation
- Heart Failure/etiology
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/therapy
- Heart Rate
- Hormone Replacement Therapy
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/therapy
- Infusion Pumps, Implantable
- Male
- Myocardial Contraction
- Myocardial Infarction/complications
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocardial Infarction/therapy
- Myosin Heavy Chains/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Recovery of Function
- Systole
- Time Factors
- Triiodothyronine/administration & dosage
- Triiodothyronine/blood
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/therapy
- Ventricular Myosins/genetics
Collapse
Affiliation(s)
- Kyle K Henderson
- Department of Medicine and the Cardiovascular Institute, Loyola University Medical Center, Maywood, Ill 60153, USA.
| | | | | | | | | | | |
Collapse
|
310
|
Fong GH. Regulation of angiogenesis by oxygen sensing mechanisms. J Mol Med (Berl) 2009; 87:549-60. [PMID: 19288062 DOI: 10.1007/s00109-009-0458-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2009] [Revised: 02/25/2009] [Accepted: 02/26/2009] [Indexed: 12/26/2022]
Abstract
The choices for blood vessels to undergo angiogenesis or stay quiescent are mostly determined by the status of tissue oxygenation. A major link between tissue hypoxia and active angiogenesis is the accumulation of hypoxia-inducible factor (HIF)-alpha subunits which play a major role in the transcriptional activation of genes encoding angiogenic factors. HIF-alpha abundance is negatively regulated by a subfamily of dioxygenases referred to as prolyl hydroxylase domain-containing proteins (PHDs) which use O(2) as a substrate to hydroxylate HIF-alpha subunits and hence tag them for rapid degradation. Under hypoxic conditions, HIF-alpha subunits accumulate due to reduced hydroxylation efficiency and form transcriptionally active heterodimers with HIF-1ss to activate the expression of angiogenic factors and other proteins important for cellular adaptation to hypoxia. Angiogenesis is regulated by a combination of at least two different mechanisms. The paracrine mechanism is mediated by non-endothelial expression of angiogenic factors such as vascular endothelial growth factor (VEGF)-A, which in turn interact with endothelial cell surface receptors to initiate angiogenic activities. In the autocrine mechanism, endothelial cell themselves are induced to express VEGF-A, which collaborate with the paracrine mechanism to support angiogenesis and protect vascular integrity. Because of critical roles of PHDs and HIFs in regulating angiogenic activities, studies are underway to assess their candidacy as targets for angiogenesis therapies.
Collapse
Affiliation(s)
- Guo-Hua Fong
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, 06030, USA.
| |
Collapse
|
311
|
|
312
|
Testa U, Pannitteri G, Condorelli GL. Vascular endothelial growth factors in cardiovascular medicine. J Cardiovasc Med (Hagerstown) 2009; 9:1190-221. [PMID: 19001927 DOI: 10.2459/jcm.0b013e3283117d37] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The discovery of vascular endothelial growth factors (VEGFs) and their receptors has considerably improved the understanding of the development and function of endothelial cells. Each member of the VEGF family appears to have a specific function: VEGF-A induces angiogenesis (i.e. growth of new blood vessels from preexisting ones), placental growth factor mediates both angiogenesis and arteriogenesis (i.e. the formation of collateral arteries from preexisting arterioles), VEGF-C and VEGF-D act mainly as lymphangiogenic factors. The study of the biology of these endothelial growth factors has allowed a major progress in the comprehension of the genesis of the vascular system and its abnormalities observed in various pathologic conditions (atherosclerosis and coronary artery disease). The role of VEGF in the atherogenic process is still unclear, but actual evidence suggests both detrimental (development of a neoangiogenetic process within the atherosclerotic plaque) and beneficial (promotion of collateral vessel formation) effects. VEGF and other angiogenic growth factors (fibroblast growth factor), although initially promising in experimental studies and in initial phase I/II clinical trials in patients with ischemic heart disease or peripheral arterial occlusive disease, have subsequently failed to show significant therapeutic improvements in controlled clinical studies. Challenges still remain about the type or the combination of angiogenic factors to be administered, the form (protein vs. gene), the route, and the duration of administration.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Italy.
| | | | | |
Collapse
|
313
|
Hamou C, Callaghan MJ, Thangarajah H, Chang E, Chang EI, Grogan RH, Paterno J, Vial IN, Jazayeri L, Gurtner GC. Mesenchymal stem cells can participate in ischemic neovascularization. Plast Reconstr Surg 2009; 123:45S-55S. [PMID: 19182663 PMCID: PMC2878772 DOI: 10.1097/prs.0b013e318191be4a] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cells from the bone marrow contribute to ischemic neovascularization, but the identity of these cells remains unclear. The authors identify mesenchymal stem cells as a bone marrow-derived progenitor population that is able to engraft into peripheral tissue in response to ischemia. METHODS A murine model of skin ischemia was used. Bone marrow, blood, and skin were harvested at different time points and subjected to flow cytometric analysis for mesenchymal and hematopoietic markers (n = 3 to 7 per time point). Using a parabiotic model pairing donor green fluorescent protein (GFP)-positive with recipient wild-type mice, progenitor cell engraftment was examined in ischemic tissue by fluorescence microscopy, and engrafted cells were analyzed by flow cytometry for endothelial and mesenchymal markers. In vitro, the ability of both bone marrow- and adipose-derived mesenchymal stem cells to adopt endothelial characteristics was examined by analyzing (1) the ability of mesenchymal stem cells to take up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and (2) phenotypic changes of mesenchymal stem cells co-cultured with GFP-labeled endothelial cells or under hypoxic/vascular endothelial growth factor stimulation. RESULTS In vivo, the bone marrow mesenchymal stem cell population decreased significantly immediately after surgery, with subsequent engraftment of these cells in ischemic tissue. Engrafted cells lacked the panhematopoietic antigen CD45, consistent with a mesenchymal origin. In vitro, bone marrow- and adipose-derived mesenchymal stem cells took up DiI-acetylated low-density lipoprotein and Alexa Fluor lectin, and expressed endothelial markers under hypoxic conditions. CONCLUSIONS The authors' data suggest that mesenchymal precursor cells can give rise to endothelial progenitors. Consequently, cell-based therapies augmenting the mesenchymal stem cell population could represent powerful alternatives to current therapies for ischemic vascular disease.
Collapse
Affiliation(s)
- Cynthia Hamou
- Stanford, Calif. From the Department of Surgery, Division of Plastic Surgery, Stanford University School of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Abstract
Angiogenesis plays an important role in the pathophysiology of atherosclerosis and after myocardial infarction. Furthermore, angiogenesis has been the focus of many therapeutic strategies. In view of that, a direct and clear understanding of the role of these pathways in the living subject is needed. Molecular Imaging has emerged as a powerful tool to study biological processes non-invasively. In this review, evidence will be presented and discussed on the feasibility of different molecular imaging strategies to study the involvement of angiogenic pathways in the assessment of the atherosclerotic disease and as a tool to assess angiogenic therapy. Focus will be placed on those imaging modalities with the potential to be translated to clinical use.
Collapse
Affiliation(s)
- Martin Rodriguez-Porcel
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
315
|
Hu ZY, Luo NF, Liu J. The protective effects of emulsified isoflurane on myocardial ischemia and reperfusion injury in rats. Can J Anaesth 2008; 56:115-25. [DOI: 10.1007/s12630-008-9016-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 09/24/2008] [Accepted: 11/14/2008] [Indexed: 10/20/2022] Open
|
316
|
Wong W, Goehring AS, Kapiloff MS, Langeberg LK, Scott JD. mAKAP compartmentalizes oxygen-dependent control of HIF-1alpha. Sci Signal 2008; 1:ra18. [PMID: 19109240 PMCID: PMC2828263 DOI: 10.1126/scisignal.2000026] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The activity of the transcription factor hypoxia-inducible factor 1alpha (HIF-1alpha) is increased in response to reduced intracellular oxygen. Enzymes of the protein ubiquitin machinery that signal the destruction or stabilization of HIF-1alpha tightly control this transcriptional response. Here, we show that muscle A kinase-anchoring protein (mAKAP) organized ubiquitin E3 ligases that managed the stability of HIF-1alpha and optimally positioned it close to its site of action inside the nucleus. Functional experiments in cardiomyocytes showed that depletion of mAKAP or disruption of its targeting to the perinuclear region altered the stability of HIF-1alpha and transcriptional activation of genes associated with hypoxia. Thus, we propose that compartmentalization of oxygen-sensitive signaling components may influence the fidelity and magnitude of the hypoxic response.
Collapse
Affiliation(s)
- Wei Wong
- 1Howard Hughes Medical Institute and Vollum Institute, Oregon Health & Science University, 3181 S. W. Sam Jackson Park Road, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
317
|
Benndorf RA, Schwedhelm E, Gnann A, Taheri R, Kom G, Didié M, Steenpass A, Ergün S, Böger RH. Isoprostanes Inhibit Vascular Endothelial Growth Factor–Induced Endothelial Cell Migration, Tube Formation, and Cardiac Vessel Sprouting In Vitro, As Well As Angiogenesis In Vivo via Activation of the Thromboxane A
2
Receptor. Circ Res 2008; 103:1037-46. [DOI: 10.1161/circresaha.108.184036] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Isoprostanes are endogenously formed end products of lipid peroxidation. Furthermore, they are markers of oxidative stress and independent risk markers of coronary heart disease. In patients experiencing coronary heart disease, impaired angiogenesis may exacerbate insufficient blood supply of ischemic myocardium. We therefore hypothesized that isoprostanes may exert detrimental cardiovascular effects by inhibiting angiogenesis. We studied the effect of isoprostanes on vascular endothelial growth factor (VEGF)-induced migration and tube formation of human endothelial cells (ECs), and cardiac angiogenesis in vitro as well as on VEGF-induced angiogenesis in the chorioallantoic membrane assay in vivo. The isoprostanes 8-iso-PGF
2α
, 8-iso-PGE
2
, and 8-iso-PGA
2
inhibited VEGF-induced migration, tube formation of ECs, and cardiac angiogenesis in vitro, as well as VEGF-induced angiogenesis in vivo via activation of the thromboxane A
2
receptor (TBXA2R): the specific TBXA2R antagonists SQ-29548, BM 567, and ICI 192,605 but not the thromboxane A
2
synthase inhibitor ozagrel blocked the effect of isoprostanes. The isoprostane 8-iso-PGA
2
degraded into 2 biologically active derivatives in vitro, which also inhibited EC tube formation via the TBXA2R. Moreover, short hairpin RNA–mediated knockdown of the TBXA2R antagonized isoprostane-induced effects. In addition, Rho kinase inhibitor Y-27632 reversed the inhibitory effect of isoprostanes and the thromboxane A
2
mimetic U-46619 on EC migration and tube formation. Finally, the various isoprostanes exerted a synergistic inhibitory effect on EC tube formation. We demonstrate for the first time that isoprostanes inhibit angiogenesis via activation of the TBXA2R. By this mechanism, isoprostanes may contribute directly to exacerbation of coronary heart disease and to capillary rarefaction in disease states of increased oxidative stress.
Collapse
Affiliation(s)
- Ralf A. Benndorf
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Edzard Schwedhelm
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Anke Gnann
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Raihana Taheri
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Ghainsom Kom
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Michael Didié
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Anna Steenpass
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Süleyman Ergün
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| | - Rainer H. Böger
- From the Clinical Pharmacology Unit (R.A.B., E.S., A.G., R.T., G.K., M.D., A.S., R.H.B.), Institute of Experimental and Clinical Toxicology and Pharmacology, University Hospital Hamburg-Eppendorf; and Institute of Anatomy (S.E.), University Hospital Essen, Germany
| |
Collapse
|
318
|
Kässmeyer S, Plendl J, Custodis P, Bahramsoltani M. New insights in vascular development: vasculogenesis and endothelial progenitor cells. Anat Histol Embryol 2008; 38:1-11. [PMID: 18983622 DOI: 10.1111/j.1439-0264.2008.00894.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the course of new blood vessel formation, two different processes--vasculogenesis and angiogenesis--have to be distinguished. The term vasculogenesis describes the de novo emergence of a vascular network by endothelial progenitors, whereas angiogenesis corresponds to the generation of vessels by sprouting from pre-existing capillaries. Until recently, it was thought that vasculogenesis is restricted to the prenatal period. During the last decade, one of the most fascinating innovations in the field of vascular biology was the discovery of endothelial progenitor cells and vasculogenesis in the adult. This review aims at introducing the concept of adult vasculogenesis and discusses the efforts to identify and characterize adult endothelial progenitors. The different sources of adult endothelial progenitors like haematopoietic stem cells, myeloid cells, multipotent progenitors of the bone marrow, side population cells and tissue-residing pluripotent stem cells are considered. Moreover, a survey of cellular and molecular control mechanisms of vasculogenesis is presented. Recent advances in research on endothelial progenitors exert a strong impact on many different disciplines and provide the knowledge for functional concepts in basic fields like anatomy, histology as well as embryology.
Collapse
Affiliation(s)
- S Kässmeyer
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | | | | | | |
Collapse
|
319
|
Xu L, Xia J, Zhang K, Xie A. Regulation of hypoxic response elements on the expression of vascular endothelial growth factor gene transfected to rat skeletal myoblasts under hypoxic environment. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2008; 28:568-571. [PMID: 18846340 DOI: 10.1007/s11596-008-0517-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Indexed: 05/26/2023]
Abstract
The regulation of hypoxic response elements on the expression of vascular endothelial growth factor (VEGF) gene transfected to primary cultured rat skeletal myoblasts under hypoxic environment was investigated. pEGFP-C3-9HRE-CMV-VEGF vector was constructed with molecular biology technique and transfected to primary cultured rat skeletal myoblasts by lipofectamine in vitro. Gene expression of transfected myoblasts was detected by RT-PCR, Western blot and fluorescence microscope under different oxygen concentrations and different hypoxia time. The results showed that in hypoxia group, the VEGF gene bands were seen and with the decrease of oxygen concentrations and prolongation of hypoxia time, the expression of VEGF mRNA was obviously increased. Under hypoxic environment, the expression of VEGF protein in the transfected myoblasts was significantly increased. EGFP was expressed only under hypoxic environment but not under normoxic environment. It was concluded that hypoxia promoter could be constructed with HRE and regulate the expression of VEGF gene under hypoxic and normoxic environment, which could enhance the reliability of gene therapy.
Collapse
Affiliation(s)
- Lei Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | | | | | | |
Collapse
|
320
|
Cruz-Gonzalez I, Pabón P, Rodríguez-Barbero A, Martín-Moreiras J, Pericacho M, Sánchez PL, Ramirez V, Sánchez-Ledesma M, Martín-Herrero F, Jiménez-Candil J, Maree AO, Sánchez-Rodríguez A, Martín-Luengo C, López-Novoa JM. Identification of serum endoglin as a novel prognostic marker after acute myocardial infarction. J Cell Mol Med 2008; 12:955-61. [PMID: 18494936 PMCID: PMC4401139 DOI: 10.1111/j.1582-4934.2008.00156.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Endoglin is a proliferation-associated and hypoxia-inducible protein expressed in endothelial cells. The levels of soluble circulating endoglin and their prognostic significance in patients with acute myocardial infarction (AMI) are not known. In this observational prospective study serum endoglin levels were measured by ELISA in 183 AMI patients upon admission to hospital and 48 hrs later and in 72 healthy controls. Endoglin levels in AMI patients on admission were significantly lower than in healthy controls (4.25 ± 0.99 ng/ml versus 4.59 ± 0.87 ng/ml; P= 0.013), and decreased further in the first 48 hours (3.65 ± 0.76 ng/ml, P < 0.001). Upon follow-up (median 319 days), patients who died had a significantly greater decrease in serum endoglin level over the first 48 hrs than those who survived (1.03 ± 0.91 versus 0.54 ± 0.55 ng/ml; P= 0.025). Endoglin decrease was an independent predictor of short-term (30 days) (hazard ratio 2.33;95% CI = 1.27–4.23; P= 0.006) cardiovascular mortality, and also predicts overall cardiovascular mortality during the follow-up (median 319 days) in AMI patients (hazard ratio 2.13;95% CI = 1.20–3.78; P= 0.01). In conclusion, early changes in serum endoglin may predict mortality after AMI.
Collapse
|
321
|
Saeed M, Martin A, Jacquier A, Bucknor M, Saloner D, Do L, Ursell P, Su H, Kan YW, Higgins CB. Permanent coronary artery occlusion: cardiovascular MR imaging is platform for percutaneous transendocardial delivery and assessment of gene therapy in canine model. Radiology 2008; 249:560-71. [PMID: 18780824 DOI: 10.1148/radiol.2491072068] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To provide evidence that vascular endothelial growth factor (VEGF) genes delivered transendocardially with magnetic resonance (MR) imaging guidance may neovascularize or improve vascular recruitment in occlusive infarction. MATERIALS AND METHODS All experimental procedures received approval from the institutional committee on animal research. Dogs with permanent coronary artery occlusion were imaged twice (3 days after occlusion for assessment of acute infarction; a mean of 50 days after occlusion +/- 3 [standard error of the mean] for assessment of chronic infarction). A mixture of plasmid VEGF and plasmid LacZ (n = 6, treated animals) or plasmid LacZ and sprodiamide (n = 6, placebo control animals) was delivered to four sites. MR fluoroscopy was used to target and monitor delivery of genes. The effectiveness of this delivery approach was determined by using MR imaging methods to assess perfusion, left ventricular (LV) function, myocardial viability, and infarct resorption. Histologic evaluation of neovascularization was then performed. RESULTS MR fluoroscopic guidance of injectates was successful in both groups. Treated animals with chronic, but not those with acute, infarction showed the following differences compared with control animals: (a) steeper mean maximum upslope perfusion (200 sec(-1) +/- 32 vs 117 sec(-1) +/- 15, P = .02), (b) higher peak signal intensity (1667 arbitrary units +/- 100 vs 1132 arbitrary units +/- 80, P = .002), (c) increased ejection fraction (from 27.9% +/- 1.2 to 35.3% +/- 1.6, P = .001), (d) smaller infarction size (as a percentage of LV mass) at MR imaging (8.5% +/- 0.9 vs 11.3% +/- 0.9, P = .048) and triphenyltetrazolium chloride staining (9.4% +/- 1.5 vs 12.7% +/- 0.4, P = .05), and (e) higher vascular density (as number of vessels per square millimeter) at the border (430 +/- 117 vs 286 +/- 19, P = .0001) and core (307 +/- 112 vs 108 +/- 17, P = .0001). CONCLUSION The validity of plasmid VEGF gene delivered with MR fluoroscopic guidance into occlusive infarction was confirmed by neovascularization associated with improved perfusion, LV function, and infarct resorption.
Collapse
Affiliation(s)
- Maythem Saeed
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94134-0628, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
322
|
Activation of negative regulators of the hypoxia-inducible factor (HIF) pathway in human end-stage heart failure. Biochem Biophys Res Commun 2008; 376:315-20. [PMID: 18782560 DOI: 10.1016/j.bbrc.2008.08.152] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2008] [Accepted: 08/26/2008] [Indexed: 01/18/2023]
Abstract
The hypoxia-inducible transcription factor HIF is induced early in acute myocardial ischemia in humans, but it is unknown whether this activation of HIF persists during chronic heart failure. The HIF system was characterized in left ventricular myocardia from 18 explanted failing hearts and 11 non-failing donor hearts by quantitative RT-PCR and Western analysis. HIF-1alpha mRNA levels were significantly decreased while its natural antisense transcript aHIF was nearly twofold higher (p<0.01) in failing myocardia than in control hearts. Moreover, compared to donor hearts a significantly increased expression of HIF-3alpha, which may act as a competitive inhibitor of HIF-1/2alpha activity, and PHD3, which upon hydroxylation of prolyl residues directs HIF-alpha subunits towards proteasomal degradation, was observed in the failing myocardium. Although negative regulators of HIF were induced, the HIF pathway obviously remains activated in chronic human heart failure, because prototype HIF target genes, such as ABCG2, VEGF, and BNIP3, were significantly induced.
Collapse
|
323
|
Rapid stabilization of vulnerable carotid plaque within 1 month of pitavastatin treatment in patients with acute coronary syndrome. J Cardiovasc Pharmacol 2008; 51:365-71. [PMID: 18427279 DOI: 10.1097/fjc.0b013e318165dcad] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We determined time course of stabilization of echolucent carotid plaques by statin therapy in patients with acute coronary syndrome (ACS). Treatment with 4 mg/d pitavastatin (n = 33) or placebo (n = 32) was initiated within 3 days after onset of ACS in 65 patients with echolucent carotid plaque. Vulnerable carotid plaques were assessed by measuring plaque echolucency using carotid ultrasound with integrated backscatter (IBS) analysis before and 1 month after treatment in all patients. The calibrated IBS value (intima-media IBS value minus adventia IBS) of vulnerable carotid plaques favorably changed at 1 month after treatment in both groups, but the echolucency at 1 month improved more in the pitavastatin than in the placebo group (pitavastatin group: -18.7 +/- 3.3 dB at pretreatment versus -12.7 +/- 2.3 dB at 1 month *P < 0.001; placebo: -19.0 +/- 3.5 dB versus -16.9 +/- 3.2 dB, P < 0.05, *P < 0.01 versus the value at 1 month in placebo group). Levels of CRP, VEGF, and TNFalpha at 1 month were significantly lower in pitavastatin than placebo group. In conclusion, pitavastatin improved carotid plaque echolucency within 1 month of therapy in patients with ACS, in association with decrease in the inflammatory biomarkers related to vulnerable plaques.
Collapse
|
324
|
Hypoxia-inducible factor-1alpha induces the coronary collaterals for coronary artery disease. Coron Artery Dis 2008; 19:173-9. [PMID: 18418234 DOI: 10.1097/mca.0b013e3282fa4b2c] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Marked variability exists in coronary artery collaterals in patients with ischemic heart disease. Multiple factors are thought to play a role in collateral development; however, the contribution of hypoxia inducible factor-1alpha (HIF-1alpha), which is a transcriptional activator that functions as a master regulator of oxygen homeostasis, is not completely clear. It could play an important role in modulating collateral development. OBJECTIVE The objective of this study is to investigate the changes and significance of expression of HIF-1alpha in patients with coronary artery collaterals. METHODS Collateral vessels were determined in 98 patients with >or=70% narrowing of at least one coronary artery without earlier revascularization, 42 patients with coronary artery collaterals and 56 patients with no coronary artery collaterals. Extent of collaterals was expressed as scores according to the Rentrop scoring system. Another 50 cases with normal coronary arteries were selected as control. The levels of HIF-1alpha protein expression in monocyte and lymphocyte in the participants were tested by immunohistochemistry (IHC) and western blot; mRNA levels were measured using reverse transcriptase PCR technique. RESULTS Compared with the control with normal coronary artery, the patients had higher expression of HIF-1alpha protein tested by IHC and western blot (52.6+/-10.2 vs. 13.7+/-6.2 by IHC, 50.8+/-4.5 vs. 6.5+/-1.8 by western blot); furthermore, significantly higher HIF-1alpha expression was observed in patients with collaterals compared with patients with no collaterals (81.5+/-11.8 vs. 20.7+/-9.4 by IHC; 87.2+/-6.5 vs. 9.5+/-1.4 by western blot). On the transcriptional levels of HIF-1alpha, the result was the same as the protein, there was significant difference of HIF-1alpha between the three groups. The patients with collaterals were the highest (127.3+/-23.9), followed by patients with no collaterals (35.7+/-12.3), and the control were the lowest (23.5+/-9.3). A highly positive correlation was observed between the expression/transcription of HIF-1alpha and collateral score (P<0.01, IHC: r1=0.78, reverse transcriptase PCR: r2=0.69, western blot: r3=0.84). CONCLUSION These data suggest that higher inductions of HIF-1alpha are associated with coronary collaterals, thus implying that HIF-1alpha may promote coronary collateral formation. Detection of HIF-1alpha expression might be helpful to predict prognosis of patients with coronary artery disease.
Collapse
|
325
|
Abstract
Angiogenesis is the biologic process of forming new blood vessels and is being investigated as an innovative therapeutic approach to help manage ischemic heart disease and peripheral vascular disease. Research studies have identified various angiogenic growth factors and progenitor cells that can enhance new blood vessel formation. Preclinical investigations in animal models have explored the potential use of growth factors with and without progenitor cells to treat myocardial ischemia. The results of clinical trials with growth factor infusions and gene therapy techniques to enhance growth factor production have shown some promise, but therapeutic angiogenesis remains at an early stage of development.
Collapse
|
326
|
Abstract
Myocardial infarction is the most common cause of cardiac injury and results in acute loss of a large number of myocardial cells. Because the heart has negligible regenerative capacity, cardiomyocyte death triggers a reparative response that ultimately results in formation of a scar and is associated with dilative remodeling of the ventricle. Cardiac injury activates innate immune mechanisms initiating an inflammatory reaction. Toll-like receptor-mediated pathways, the complement cascade and reactive oxygen generation induce nuclear factor (NF)-kappaB activation and upregulate chemokine and cytokine synthesis in the infarcted heart. Chemokines stimulate the chemotactic recruitment of inflammatory leukocytes into the infarct, while cytokines promote adhesive interactions between leukocytes and endothelial cells, resulting in transmigration of inflammatory cells into the site of injury. Monocyte subsets play distinct roles in phagocytosis of dead cardiomyocytes and in granulation tissue formation through the release of growth factors. Clearance of dead cells and matrix debris may be essential for resolution of inflammation and transition into the reparative phase. Transforming growth factor (TGF)-beta plays a crucial role in cardiac repair by suppressing inflammation while promoting myofibroblast phenotypic modulation and extracellular matrix deposition. Myofibroblast proliferation and angiogenesis result in formation of highly vascularized granulation tissue. As the healing infarct matures, fibroblasts become apoptotic and a collagen-based matrix is formed, while many infarct neovessels acquire a muscular coat and uncoated vessels regress. Timely resolution of the inflammatory infiltrate and spatial containment of the inflammatory and reparative response into the infarcted area are essential for optimal infarct healing. Targeting inflammatory pathways following infarction may reduce cardiomyocyte injury and attenuate adverse remodeling. In addition, understanding the role of the immune system in cardiac repair is necessary in order to design optimal strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Section of Cardiovascular Sciences, Baylor College of Medicine, One Baylor Plaza BCM620, Houston, TX 77030, United States.
| |
Collapse
|
327
|
Roncalli J, Tongers J, Renault MA, Losordo DW. Biological approaches to ischemic tissue repair: gene- and cell-based strategies. Expert Rev Cardiovasc Ther 2008; 6:653-68. [PMID: 18510483 DOI: 10.1586/14779072.6.5.653] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gene therapy is a potential therapeutic strategy for treatment of ischemic vascular diseases; however, the clinical application of gene therapy has met some anticipated challenges. Recent randomized, controlled trials suggest that patients with cardiovascular disease may also benefit from cell-based therapies, and the optimal treatment regimen may combine both approaches to take advantage of potential synergy between the underlying therapeutic mechanisms. This review discusses recent research into both gene and cell therapy and considers the potential application of a combined treatment approach for cardiovascular and cerebrovascular ischemic diseases.
Collapse
Affiliation(s)
- Jerome Roncalli
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | |
Collapse
|
328
|
Zhao W, Han Q, Lin H, Gao Y, Sun W, Zhao Y, Wang B, Chen B, Xiao Z, Dai J. Improved neovascularization and wound repair by targeting human basic fibroblast growth factor (bFGF) to fibrin. J Mol Med (Berl) 2008; 86:1127-38. [DOI: 10.1007/s00109-008-0372-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 04/22/2008] [Accepted: 05/19/2008] [Indexed: 01/12/2023]
|
329
|
Novotny NM, Ray R, Markel TA, Crisostomo PR, Wang M, Wang Y, Meldrum DR. Stem cell therapy in myocardial repair and remodeling. J Am Coll Surg 2008; 207:423-34. [PMID: 18722949 DOI: 10.1016/j.jamcollsurg.2008.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 04/04/2008] [Accepted: 04/07/2008] [Indexed: 01/01/2023]
Affiliation(s)
- Nathan M Novotny
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | |
Collapse
|
330
|
Spyridopoulos I, Erben Y, Brummendorf TH, Haendeler J, Dietz K, Seeger F, Kissel CK, Martin H, Hoffmann J, Assmus B, Zeiher AM, Dimmeler S. Telomere Gap Between Granulocytes and Lymphocytes Is a Determinant for Hematopoetic Progenitor Cell Impairment in Patients With Previous Myocardial Infarction. Arterioscler Thromb Vasc Biol 2008; 28:968-74. [DOI: 10.1161/atvbaha.107.160846] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objective—
We have previously demonstrated that ischemic cardiomyopathy is associated with selective impairment of progenitor cell function in the bone marrow and in the peripheral blood, which may contribute to an unfavorable left ventricular remodeling process.
Methods and Results—
With this study, we intended to identify the influence of telomere length on bone marrow functionality in 50 patients with coronary artery disease (CAD) and previous myocardial infarction. Mean telomere length (mTL) was measured simultaneously in peripheral blood leukocytes and mononuclear bone marrow cells (BMC), using the flow-FISH method. Telomere erosion already occurred at the bone marrow level, whereby age (39 bp/yr,
P
=0.025) and the number of affected vessels (434 bp/vessel,
P
=0.029) were the only independent predictors. Lymphocytes demonstrated significant TL shortening between BMCs and peripheral blood in CAD patients (−1011±897 bp) as opposed to an increase in a young control group (+235±459 bp,
P
<0.001). SDF- and VEGF-specific migration of BMCs correlated with mTL of lymphocytes (
r
=0.42,
P
<0.001) and was significantly reduced in CAD patients. Finally, the telomere length difference between granulocytes and lymphocytes was the most determinant for telomere-associated bone marrow impairment (
P
<0.001).
Conclusion—
In patients with CAD, telomere shortening of BMCs is dependent on both age and the extent of CAD and correlates with bone marrow cell functionality.
Collapse
Affiliation(s)
- Ioakim Spyridopoulos
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Young Erben
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Tim H. Brummendorf
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Judith Haendeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Klaus Dietz
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Florian Seeger
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Christine K. Kissel
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Hans Martin
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Jedrzej Hoffmann
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Birgit Assmus
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| |
Collapse
|
331
|
Arab A, Kuemmerer K, Wang J, Bode C, Hehrlein C. Oxygenated perfluorochemicals improve cell survival during reoxygenation by pacifying mitochondrial activity. J Pharmacol Exp Ther 2008; 325:417-24. [PMID: 18305017 DOI: 10.1124/jpet.107.133710] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Perfluorochemicals (PFCs) are known to provide a unique tool for controlled uptake and delivery of oxygen. We have characterized the effects of incremental oxygen delivery on cell viability of human ischemic cardiomyocytes using chemically inert PFCs as oxygen carrier. We have found that cell viability after prolonged ischemia depends on the dose of oxygen supplementation by oxygenated (ox) PFCs during reoxygenation. Although reoxygenation with the transient addition of oxPFCs in high concentrations (2250 microMO2 in 0.4 muM PFCs) results in decreased cell viability compared with normoxic reoxygenation, cell survival increases by 30 +/- 4% after reoxygenation with moderate oxPFC concentrations (750 muM O2 in 0.1 microM PFCs). Immunoblot analysis revealed that oxPFC-supplemented reoxygenation causes marked (16-fold) deactivation of death-associated protein kinase (DAPK) signaling an increase in mitochondrial membrane potential and a decreased steady-state level of superoxide by 19 +/- 3%. Reoxygenation with oxPFCs is further responsible for a 2-fold activation of AMP-activated protein kinase (AMPK) signaling an inadequate ATP supply by oxidative phosphorylation during reoxygenation. Addition of oxPFCs stabilizes both hypoxia-inducible factor (HIF) 1-alpha and 2-alpha during reoxygenation. Overall, these results indicate that moderate doses of oxPFCs can improve cell survival during reoxygenation, causing deactivation of DAPK, up-regulation of AMPK, and HIF1-alpha and 2-alpha stabilization. These effects of oxPFCs are dose-dependent, and they lead to a stabilization of the mitochondrial membrane potential, decreased steady-state levels of superoxide, and pacification of mitochondrial activity.
Collapse
Affiliation(s)
- Amina Arab
- Department of Cardiology, University of Freiburg, Hugstetterstrasse 55, D-79106 Freiburg i. Br, Germany.
| | | | | | | | | |
Collapse
|
332
|
Psaltis PJ, Gronthos S, Worthley SG, Zannettino AC. Cellular Therapy for Cardiovascular Disease Part 2—Delivery of Cells and Clinical Experience. Clin Med Cardiol 2008. [DOI: 10.4137/117954682000200001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Peter J Psaltis
- Cardiovascular Research Centre, Royal Adelaide Hospital; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Stan Gronthos
- Division of Haematology, Institute of Medical and Veterinary Science; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Stephen G Worthley
- Cardiovascular Research Centre, Royal Adelaide Hospital; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Andrew Cw Zannettino
- Division of Haematology, Institute of Medical and Veterinary Science; Department of Medicine, University of Adelaide, South Australia, 5000
| |
Collapse
|
333
|
Chang SA, Lee EJ, Kang HJ, Zhang SY, Kim JH, Li L, Youn SW, Lee CS, Kim KH, Won JY, Sohn JW, Park KW, Cho HJ, Yang SE, Oh WI, Yang YS, Ho WK, Park YB, Kim HS. Impact of myocardial infarct proteins and oscillating pressure on the differentiation of mesenchymal stem cells: effect of acute myocardial infarction on stem cell differentiation. Stem Cells 2008; 26:1901-12. [PMID: 18403756 DOI: 10.1634/stemcells.2007-0708] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cell transplantation in acute myocardial infarction (AMI) has emerged as a promising therapeutic option. We evaluated the impact of AMI on mesenchymal stem cell (MSC) differentiation into cardiomyocyte lineage. Cord blood-derived human MSCs were exposed to in vitro conditions simulating in vivo environments of the beating heart with acute ischemia, as follows: (a) myocardial proteins or serum obtained from sham-operated rats, and (b) myocardial proteins or serum from AMI rats, with or without application of oscillating pressure. Expression of cardiac-specific markers on MSCs was greatly induced by the infarcted myocardial proteins, compared with the normal proteins. It was also induced by application of oscillating pressure to MSCs. Treatment of MSCs with infarcted myocardial proteins and oscillating pressure greatly augmented expression of cardiac-specific genes. Such expression was blocked by inhibitor of transforming growth factor beta(1) (TGF-beta(1)) or bone morphogenetic protein-2 (BMP-2). In vitro cellular and electrophysiologic experiments showed that these differentiated MSCs expressing cardiomyocyte-specific markers were able to make a coupling with cardiomyocytes but not to selfbeat. The pathophysiologic significance of in vitro results was confirmed using the rat AMI model. The protein amount of TGF-beta(1) and BMP-2 in myocardium of AMI was significantly higher than that in normal myocardium. When MSCs were transplanted to the heart and analyzed 8 weeks later, they expressed cardiomyocyte-specific markers, leading to improved cardiac function. These in vitro and in vivo results suggest that infarct-related biological and physical factors in AMI induce commitment of MSCs to cardiomyocyte-like cells through TGF-beta/BMP-2 pathways.
Collapse
Affiliation(s)
- Sung-A Chang
- National Research Laboratory for Cardiovascular Stem Cells, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
334
|
Rodriguez-Porcel M, Cai W, Gheysens O, Willmann JK, Chen K, Wang H, Chen IY, He L, Wu JC, Li ZB, Mohamedali KA, Kim S, Rosenblum MG, Chen X, Gambhir SS. Imaging of VEGF receptor in a rat myocardial infarction model using PET. J Nucl Med 2008; 49:667-73. [PMID: 18375924 PMCID: PMC2853914 DOI: 10.2967/jnumed.107.040576] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Myocardial infarction (MI) leads to left ventricular (LV) remodeling, which leads to the activation of growth factors such as vascular endothelial growth factor (VEGF). However, the kinetics of a growth factor's receptor expression, such as VEGF, in the living subject has not yet been described. We have developed a PET tracer (64Cu-DOTA-VEGF121 [DOTA is 1,4,7,10-tetraazadodecane-N,N',N'',N'''-tetraacetic acid]) to image VEGF receptor (VEGFR) expression after MI in the living subject. METHODS In Sprague-Dawley rats, MI was induced by ligation of the left coronary artery and confirmed by ultrasound (n = 8). To image and study the kinetics of VEGFRs, 64Cu-DOTA-VEGF121 PET scans were performed before MI induction (baseline) and on days 3, 10, 17, and 24 after MI. Sham-operated animals served as controls (n = 3). RESULTS Myocardial origin of the 64Cu-DOTA-VEGF121 signal was confirmed by CT coregistration and autoradiography. VEGFR specificity of the 64Cu-DOTA-VEGF121 probe was confirmed by in vivo use of a 64Cu-DOTA-VEGFmutant. Baseline myocardial uptake of 64Cu-DOTA-VEGF121 was minimal (0.30 +/- 0.07 %ID/g [percentage injected dose per gram of tissue]); it increased significantly after MI (day 3, 0.97 +/- 0.05 %ID/g; P < 0.05 vs. baseline) and remained elevated for 2 wk (up to day 17 after MI), after which time it returned to baseline levels. CONCLUSION We demonstrate the feasibility of imaging VEGFRs in the myocardium. In summary, we imaged and described the kinetics of 64Cu-DOTA-VEGF121 uptake in a rat model of MI. Studies such as the one presented here will likely play a major role when studying pathophysiology and assessing therapies in different animal models of disease and, potentially, in patients.
Collapse
Affiliation(s)
- Martin Rodriguez-Porcel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Weibo Cai
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Olivier Gheysens
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Jürgen K. Willmann
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Kai Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Hui Wang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Ian Y. Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
| | - Lina He
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Joseph C. Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
- Department of Cardiology, Stanford University, Stanford, California
| | - Zi-bo Li
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Khalid A. Mohamedali
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Sehoon Kim
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Michael G. Rosenblum
- Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, Texas
| | - Xiaoyuan Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Division of Nuclear Medicine, Stanford University, Stanford, California
- Department of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
335
|
Kalinowski L, Dobrucki LW, Meoli DF, Dione DP, Sadeghi MM, Madri JA, Sinusas AJ. Targeted imaging of hypoxia-induced integrin activation in myocardium early after infarction. J Appl Physiol (1985) 2008; 104:1504-12. [PMID: 18356482 DOI: 10.1152/japplphysiol.00861.2007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The alphavbeta3-integrin is expressed in angiogenic vessels in response to hypoxia and represents a potential novel target for imaging myocardial angiogenesis. This study evaluated the feasibility of noninvasively tracking hypoxia-induced alphavbeta3-integrin activation within the myocardium as a marker of angiogenesis early after myocardial infarction. Acute myocardial infarction was produced by coronary artery occlusion in rodent and canine studies. A novel (111)In-labeled radiotracer targeted at the alphavbeta3-integrin ((111)In-RP748) was used to localize regions of hypoxia-induced angiogenesis early after infarction. In rodent studies, the specificity of (111)In-RP748 for alphavbeta3-integrin was confirmed with a negative control compound ((111)In-RP790), and regional uptake of these compounds correlated with (201)Tl perfusion and a (99m)Tc-labeled nitroimidazole (BRU59-21), which was used as a quantitative marker of myocardial hypoxia. The ex vivo analysis demonstrated that only (111)In-RP748 was selectively retained in infarcted regions with reduced (201)Tl perfusion and correlated with uptake of BRU59-21. In canine studies, myocardial uptake of (111)In-RP748 was assessed using in vivo single-photon-emission computed tomography (SPECT), ex vivo planar imaging, and gamma well counting of myocardial tissue and correlated with (99m)Tc-labeled 2-methoxy-2-methyl-propyl-isonitrile ((99m)Tc-sestamibi) perfusion. Dual-radiotracer in vivo SPECT imaging of (111)In-RP748 and (99m)Tc-sestamibi provided visualization of (111)In-RP748 uptake within the infarct region, which was confirmed by ex vivo planar imaging of excised myocardial slices. Myocardial (111)In-RP748 retention was associated with histological evidence of alphavbeta3-integrin expression/activation in the infarct region. (111)In-RP748 imaging provides a novel noninvasive approach for evaluation of hypoxia-induced alphavbeta3-integrin activation in myocardium early after infarction and may prove useful for directing and evaluating angiogenic therapies in patients with ischemic heart disease.
Collapse
Affiliation(s)
- Leszek Kalinowski
- Experimental Nuclear Cardiology Laboratory, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8017, USA
| | | | | | | | | | | | | |
Collapse
|
336
|
Fong GH. Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis 2008; 11:121-40. [PMID: 18327686 DOI: 10.1007/s10456-008-9107-3] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Accepted: 02/25/2008] [Indexed: 12/18/2022]
Abstract
Angiogenesis is mostly an adaptive response to tissue hypoxia, which occurs under a wide variety of situations ranging from embryonic development to tumor growth. In general, angiogenesis is dependent on the accumulation of hypoxia inducible factors (HIFs), which are heterodimeric transcription factors of alpha and beta subunits. Under normoxia, HIF heterodimers are not abundantly present due to oxygen dependent hydroxylation, polyubiquitination, and proteasomal degradation of alpha subunits. Under hypoxia, however, alpha subunits are stabilized and form heterodimers with HIF-1beta which is not subject to oxygen dependent regulation. The accumulation of HIFs under hypoxia allows them to activate the expression of many angiogenic genes and therefore initiates the angiogenic process. In recent years, however, it has become clear that various other mechanisms also participate in fine tuning angiogenesis. In this review, I discuss the relationship between hypoxia and angiogenesis under five topics: (1) regulation of HIF-alpha abundance and activity by oxygen tension and other conditions including oxygen independent mechanisms; (2) hypoxia-regulated expression of angiogenic molecules by HIFs and other transcription factors; (3) responses of vascular cells to hypoxia; (4) angiogenic phenotypes due to altered HIF signaling in mice; and (5) role of the HIF pathway in pathological angiogenesis. Studies discussed under these topics clearly indicate that while mechanisms of oxygen-regulated HIF-alpha stability provide exciting opportunities for the development of angiogenesis or anti-angiogenesis therapies, it is also highly important to consider various other mechanisms for the optimization of these procedures.
Collapse
Affiliation(s)
- Guo-Hua Fong
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3501, USA.
| |
Collapse
|
337
|
Obata JE, Kugiyama K. Reply. J Am Coll Cardiol 2008. [DOI: 10.1016/j.jacc.2007.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
338
|
Hypoxia-inducible factor-dependent degeneration, failure, and malignant transformation of the heart in the absence of the von Hippel-Lindau protein. Mol Cell Biol 2008; 28:3790-803. [PMID: 18285456 DOI: 10.1128/mcb.01580-07] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hypoxia-inducible transcription factor 1 (HIF-1) and HIF-2alpha regulate the expression of an expansive array of genes associated with cellular responses to hypoxia. Although HIF-regulated genes mediate crucial beneficial short-term biological adaptations, we hypothesized that chronic activation of the HIF pathway in cardiac muscle, as occurs in advanced ischemic heart disease, is detrimental. We generated mice with cardiac myocyte-specific deletion of the von Hippel-Lindau protein (VHL), an essential component of an E3 ubiquitin ligase responsible for suppressing HIF levels during normoxia. These mice were born at expected frequency and thrived until after 3 months postbirth, when they developed severe progressive heart failure and premature death. VHL-null hearts developed lipid accumulation, myofibril rarefaction, altered nuclear morphology, myocyte loss, and fibrosis, features seen for various forms of human heart failure. Further, nearly 50% of VHL(-/-) hearts developed malignant cardiac tumors with features of rhabdomyosarcoma and the capacity to metastasize. As compelling evidence for the mechanistic contribution of HIF-1alpha, the concomitant deletion of VHL and HIF-1alpha in the heart prevented this phenotype and restored normal longevity. These findings strongly suggest that chronic activation of the HIF pathway in ischemic hearts is maladaptive and contributes to cardiac degeneration and progression to heart failure.
Collapse
|
339
|
Role of hypoxia-inducible factor in cell survival during myocardial ischemia-reperfusion. Cell Death Differ 2008; 15:686-90. [PMID: 18259200 DOI: 10.1038/cdd.2008.13] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Hypoxia-inducible factor (HIF) is the principal transcription factor involved in the regulation of transcriptional responses to hypoxia. During hypoxia, HIF-alpha levels accumulate and trigger an increase in expression of genes involved in glycolysis, glucose metabolism, mitochondrial function, cell survival, apoptosis, and resistance to oxidative stress. In this regard, HIF activation plays an essential role in triggering cellular protection and metabolic alterations from the consequences of oxygen deprivation. This suggests that HIF activation should confer protection against ischemia-reperfusion (I/R) injury, although this protection might require HIF activation before the onset of lethal ischemia. Studies using enhanced expression of HIF-1alpha suggest that its upregulation may be a beneficial therapeutic modality in the treatment or prevention of ischemic injury. HIF-regulated gene expression may mediate the late phase of preconditioning, and constitutive HIF activity may influence the expression of genes that are required for the cell to be able to respond to acute preconditioning. This article reviews the current literature on the role of HIF in balancing protection and cell death in the face of ischemia and I/R injury.
Collapse
|
340
|
Luo D, Yang D, Lan X, Li K, Li X, Chen J, Zhang Y, Xiao RP, Han Q, Cheng H. Nuclear Ca2+ sparks and waves mediated by inositol 1,4,5-trisphosphate receptors in neonatal rat cardiomyocytes. Cell Calcium 2008; 43:165-74. [PMID: 17583790 PMCID: PMC2266086 DOI: 10.1016/j.ceca.2007.04.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 04/16/2007] [Accepted: 04/29/2007] [Indexed: 11/21/2022]
Abstract
Dynamic nuclear Ca(2+) signals play pivotal roles in diverse cellular functions including gene transcription, cell growth, differentiation, and apoptosis. Here we report a novel nuclear Ca(2+) regulatory mechanism mediated by inositol 1,4,5-trisphosphate receptors (IP(3)Rs) around the nucleus in developing cardiac myocytes. Activation of IP(3)Rs by alpha(1)-adrenergic receptor (alpha(1)AR) stimulation or by IP(3) application (in saponin-permeabilized cells) increases Ca(2+) spark frequency preferentially in the region around the nucleus in neonatal rat ventricular myocytes. A nuclear enrichment of IP(3)R distribution supports the higher responsiveness of Ca(2+) release in this particular region. Strikingly, we observed "nuclear Ca(2+)waves" that engulf the entire nucleus without spreading into the bulk cytosol. alpha(1)AR stimulation enhances the occurrence of nuclear Ca(2+) waves and confers them the ability to trigger cytosolic Ca(2+) waves via IP(3)R-dependent pathways. This finding accounts, at least partly, for a profound frequency-dependent modulation of global Ca(2+) oscillations during alpha(1)AR stimulation. Thus, IP(3)R-mediated Ca(2+) waves traveling in the nuclear region provide active, autonomous regulation of nuclear Ca(2+) signaling, which provides for not only the local signal transduction, but also a pacemaker to drive global Ca(2+) transient in the context of alpha(1)AR stimulation in developing cardiac myocytes.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Animals
- Animals, Newborn
- Calcium Signaling/physiology
- Cell Membrane Permeability
- Inositol 1,4,5-Trisphosphate Receptors/drug effects
- Inositol 1,4,5-Trisphosphate Receptors/physiology
- Microscopy, Confocal
- Myocytes, Cardiac/physiology
- Nuclear Envelope/physiology
- Phenylephrine/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/physiology
Collapse
Affiliation(s)
- Dali Luo
- Department of Pharmacology, School of Chemical Biology & Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
341
|
Decreased small arterial compliance with increased serum vascular endothelial growth factor-A and circulating endothelial progenitor cell in dilated cardiomyopathy. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200802020-00007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
342
|
Endothelial precursor cells. ACTA ACUST UNITED AC 2008; 3:218-25. [PMID: 17917135 DOI: 10.1007/s12015-007-0007-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/11/2022]
Abstract
The discovery and subsequent characterization of endothelial precursor cells (EPCs) has stimulated interest in their potential use in older persons. Understanding the mechanisms that underlie EPC availability and function has important clinical implications for this age group. In this review, we discuss aspects of EPCs that are relevant to their role in angiogenesis and cardiovascular disease. We then review the limited data on features of EPCs that are known to be altered in aging and might better define their clinical utility in older persons.
Collapse
|
343
|
In vivo hepatocyte growth factor gene transfer reduces myocardial ischemia-reperfusion injury through its multiple actions. J Card Fail 2008; 13:874-83. [PMID: 18068622 DOI: 10.1016/j.cardfail.2007.07.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 06/27/2007] [Accepted: 07/09/2007] [Indexed: 11/22/2022]
Abstract
BACKGROUND Hepatocyte growth factor (HGF) is reported to protect the heart against ischemia-reperfusion injury. However, whether in vivo adenovirus-mediated HGF gene transfer before ischemia is protective against ischemia-reperfusion and its precise mechanisms are still unknown. METHODS AND RESULTS By using a rabbit model of ischemia-reperfusion injury, we demonstrate that HGF gene transfer is cardioprotective through its multiple beneficial actions, such as angiogenesis, Bcl-2 overexpression, and decreasing hydroxyl radicals, deoxyuride-5'-triphosphate biotin nick end labeling (TUNEL)-positive myocytes, and fibrotic area. After HGF gene transfer, the rabbits underwent 30 minutes of coronary occlusion and 30 minutes, 4 hours, 48 hours, and 14 days of reperfusion. The infarct size at 48 hours of reperfusion was significantly reduced in the HGF group (13.4% +/- 2.3%) compared with that in the LacZ group (36.5% +/- 2.0%) and saline group (40.3% +/- 3.2%). At 14 days of reperfusion, HGF gene transfer improved left ventricular ejection fraction and fractional shortening, reduced the fibrotic area, and increased the capillary density in the risk area. At 4 hours of reperfusion, Bcl-2 protein was overexpressed and the incidence of TUNEL-positive myocytes was significantly decreased in the risk area in the HGF group compared with the LacZ and saline groups. The myocardial interstitial 2,5-dihydroxybenzoic acid level, an indicator of hydroxyl radical, increased during 30 minutes of ischemia and 30 minutes of reperfusion in the LacZ and saline groups, and was significantly inhibited in the HGF group. CONCLUSION HGF gene therapy may be a novel therapeutic strategy against unstable angina pectoris or severe angina pectoris, which may progress to acute myocardial infarction.
Collapse
|
344
|
Wang F, Keimig T, He Q, Ding J, Zhang Z, Pourabdollah-Nejad S, Yang XP. Augmented healing process in female mice with acute myocardial infarction. ACTA ACUST UNITED AC 2008; 4:230-47. [PMID: 18022590 DOI: 10.1016/s1550-8579(07)80043-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND It is well established that premenopausal women are protected from cardiovascular disease. This gender difference in favor of females is also demonstrated in animal studies. Our research group previously found that female mice had much lower incidence of cardiac rupture and mortality than did males during the acute phase of myocardial infarction (MI); however, the mechanisms responsible for such protection are not fully understood. OBJECTIVE The aim of this study was to determine whether the favorable cardiac effect observed in female mice with MI is due to an augmented healing process that includes less inflammation, reduced matrix degradation, and enhanced neovascularization. METHODS Twelve-week-old male and female C57BL/6J mice were subjected to MI by ligating the left anterior descending coronary artery and then euthanized at 1, 4, 7, or 14 days post-MI. Inflammatory cell infiltration and myofibroblast transformation, matrix metalloproteinase (MMP)-2 and MMP-9 activity, tissue inhibitor of metalloproteinase (TIMP)-I expression, and neovascularization were examined by immunohistochemistry, zymography, Western blot, and laser scanning confocal microscopy, respectively. Cardiac function was evaluated by echocardiography on day 14. RESULTS We found that: (1) neutrophil infiltration during the early phase of MI (1-4 days) was much lower in females than in males and was associated with lower MMP-9 activity and higher TIMP-1 protein expression, indicating less-exaggerated inflammation and extracellular matrix degradation in females; (2) myofibroblast transformation, as indicated by expression of alpha-smooth muscle actin, was significantly greater in females than in males at day 7 of MI (P<0.05), indicating facilitated collagen deposition and scar formation; and (3) neovascularization (vascular area in the infarct border) was markedly increased in females, and was associated with better preserved cardiac function and less left ventricular dilatation. CONCLUSION Our data suggest that less-exaggerated early inflammation and augmented reparative fibrotic response, indicated by enhanced myofibroblast transformation, may contribute greatly to low rupture rates in females during the acute and subacute phases of MI, whereas enhanced neovascularization may lead to better preserved cardiac function post-MI.
Collapse
Affiliation(s)
- Fangfei Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, and Department of Biology, Wayne State University, Detroit, Michigan 48202-2689, USA
| | | | | | | | | | | | | |
Collapse
|
345
|
Duan HF, Wang H, Yi J, Liu HJ, Zhang QW, Li LB, Zhang T, Lu Y, Wu CT, Wang LS. Adenoviral gene transfer of sphingosine kinase 1 protects heart against ischemia/reperfusion-induced injury and attenuates its postischemic failure. Hum Gene Ther 2008; 18:1119-28. [PMID: 17939750 DOI: 10.1089/hum.2007.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sphingosine kinase 1 (SPK1) has been identified as a central mediator of ischemia preconditioning and plays a protective role in ischemia/reperfusion (I/R)-induced cardiomyocyte death. In the present study, we investigated the protective effect of adenovirus-mediated SPK1 gene (Ad-SPK1) transfer on I/R-induced cardiac injury, and evaluated its therapeutic action on postinfarction heart failure. Cardiac SPK1 activity was increased about 5-fold by injection of Ad-SPK1, compared with injection of adenovirus carrying the green fluorescent protein gene (Ad-GFP). A more potent performance and a lower incidence of arrhythmia were observed in Ad-SPK1-injected hearts during the reperfusion period, compared with Ad-GFP-injected hearts. An enzymatic activity assay showed that creatine kinase release was also less in Ad-SPK1-injected hearts. To investigate the therapeutic action of the SPK1 gene on postischemic heart failure, the left anterior descending branch of the coronary artery in Wistar rats was ligated after direct intramyocardial injection of Ad-SPK1 or Ad-GFP as a control. Ad-SPK1 injection significantly preserved cardiac systolic and diastolic function, as evidenced by left ventricular (LV) systolic pressure, LV end-diastolic pressure, and peak velocity of contraction (dP/dt). The LV morphometric parameters of Ad-SPK1-treated animals were also preserved. In addition, SPK1 gene delivery significantly enhanced angiogenesis and reduced fibrosis. These results demonstrate that adenovirus-mediated SPK1 gene transfer could efficiently prevent I/R-induced myocardial injury and attenuate postischemic heart failure. Thus, SPK1 gene delivery would be a novel strategy for the treatment of coronary heart disease.
Collapse
Affiliation(s)
- Hai-Feng Duan
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing 100850, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
Ayerden Ebinç F, Haksun E, Ulver DB, Koç E, Erten Y, Reis Altok K, Bali M, Turgay A, Sindel S. The relationship between vascular endothelial growth factor (VEGF) and microalbuminuria in patients with essential hypertension. Intern Med 2008; 47:1511-6. [PMID: 18758126 DOI: 10.2169/internalmedicine.47.1122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The existence of microalbuminuria (MAU) in patients with essential hypertension is a strong indicator of microvascular damage. Although endothelial dysfunction and increased vascular permeability both have a role in the development of MAU, its ethiopathogenesis in hypertensive patients is not yet clearly understood. Vascular endothelial growth factor (VEGF) is the most important regulator of pathological or physiological angiogenesis and it additionally leads to increased vascular permeability. This study aims to assess the relationship of serum VEGF levels to MAU in non-complicated, newly-diagnosed essential hypertensive patients (EHs). METHODS This study included 30 newly-diagnosed EHs with MAU, 46 newly-diagnosed EHs without MAU and 46 healthy controls. None of the EHs had diabetes, renal impairment or atherosclerotic diseases. Serum VEGF levels were measured using the ELISA method. RESULTS Serum levels of VEGF were significantly higher in EHs with MAU when compared with patients without MAU (225.15+/-109.34 pg/mL versus 166.78+/-114.35 pg/mL, p: 0.04) or controls (225.15+/-109.34 pg/mL versus 144.91+/-96.60 pg/mL, p: 0.007). On the other hand, no significant difference was observed between the non-MAU and control groups. In the univariate analysis, serum levels of VEGF, were positively correlated with systolic blood pressure (R: 0.253 p: 0.001), diastolic blood pressure (R: 0.162 p: 0.04), mean arterial pressure (R: 0.239 p: 0.002), creatinine clearance (R: 0.172 p: 0.04) and MAU (R: 0.338 p: 0.002). In the multiple linear regression analysis, VEGF levels were independently related to MAU (beta: 0.248, p: 0.02). CONCLUSION VEGF levels are higher in EHs in the presence of MAU. These high values may be important in the early diagnosis of vascular damage in EHs. Additionally, VEGF may increase glomerular permeability and lead to MAU in EHs.
Collapse
|
347
|
Zhu BL, Tanaka S, Ishikawa T, Zhao D, Li DR, Michiue T, Quan L, Maeda H. Forensic pathological investigation of myocardial hypoxia-inducible factor-1α, erythropoietin and vascular endothelial growth factor in cardiac death. Leg Med (Tokyo) 2008; 10:11-9. [DOI: 10.1016/j.legalmed.2007.06.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 05/29/2007] [Accepted: 06/04/2007] [Indexed: 10/23/2022]
|
348
|
Effect of modified glucose-insulin-potassium on free fatty acids, matrix metalloproteinase, and myoglobin in ST-elevation myocardial infarction. Am J Cardiol 2007; 100:1614-8. [PMID: 18036357 DOI: 10.1016/j.amjcard.2007.07.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 07/01/2007] [Accepted: 07/01/2007] [Indexed: 11/21/2022]
Abstract
Insulin has a free fatty acid (FFA)-suppressive effect, vascular endothelial growth factor (VEGF)- and matrix metalloproteinase (MMP)-lowering effect, and a potential myocardial-protective effect. Whether low-dose insulin exerts these effects in patients with acute myocardial infarction (MI) was investigated. Thirty-two patients administered thrombolytics and heparin were randomly assigned to a modified glucose-insulin-potassium (GIK) regimen (insulin 2.5 U/hour, dextrose and potassium titrated to prevent hyperglycemia) or normal saline solution and potassium (controls) for 48 hours. Plasma FFA, serum VEGF, pro-MMP-1, and myoglobin were measured at baseline and sequentially for 48 hours. FFA concentrations were increased at baseline; increased further in the first 4 hours in controls (p<or=0.008), but not in the GIK group, and were higher at 4 hours in controls compared with the GIK group (p=0.0009). VEGF decreased to 7% of baseline at 2 hours and remained suppressed in both groups (p=0.0008). Pro-MMP-1 decreased in both groups (p<0.005), but this decrease was seen earlier at 2 hours in the GIK group compared with 4 hours in controls. There was no significant increase in myoglobin at 2 hours in the GIK group, whereas there was a significant increase in controls. Mean blood glucose was 131 mg/dl in controls and 124 mg/dl in the GIK group (p=NS). In conclusion, this modified GIK regimen attenuated the increase in FFA, but did not suppress it to less than the threshold for myocardial FFA uptake. It suppressed pro-MMP-1 rapidly and decreased myoglobin, whereas heparin suppressed VEGF in patients with acute MI. This provided additional rationale for conducting a trial to assess the clinical benefits of this modified GIK regimen in patients with acute MI.
Collapse
|
349
|
Polymorphisms in hypoxia inducible factor 1 and the initial clinical presentation of coronary disease. Am Heart J 2007; 154:1035-42. [PMID: 18035072 DOI: 10.1016/j.ahj.2007.07.042] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Accepted: 07/31/2007] [Indexed: 11/22/2022]
Abstract
BACKGROUND Only some patients with coronary artery disease (CAD) develop acute myocardial infarction (MI), and emerging evidence suggests vulnerability to MI varies systematically among patients and may have a genetic component. The goal of this study was to assess whether polymorphisms in genes encoding elements of pathways mediating the response to ischemia affect vulnerability to MI among patients with underlying CAD. METHODS We prospectively identified patients at the time of their initial clinical presentation of CAD who had either an acute MI or stable exertional angina. We collected clinical data and genotyped 34 polymorphisms in 6 genes (ANGPT1, HIF1A, THBS1, VEGFA, VEGFC, VEGFR2). RESULTS The 909 patients with acute MI were significantly more likely than the 466 patients with stable angina to be male, current smokers, and hypertensive, and less likely to be taking beta-blockers or statins. Three polymorphisms in HIF1A (Pro582Ser, rs11549465; rs1087314; and Thr418Ile, rs41508050) were significantly more common in patients who presented with stable exertional angina rather than acute MI, even after statistical adjustment for cardiac risk factors and medications. The HIF-mediated transcriptional activity was significantly lower when HIF1A null fibroblasts were transfected with variant HIF1A alleles than with wild-type HIF1A alleles. CONCLUSIONS Polymorphisms in HIF1A were associated with development of stable exertional angina rather than acute MI as the initial clinical presentation of CAD.
Collapse
|
350
|
Shohet RV, Garcia JA. Keeping the engine primed: HIF factors as key regulators of cardiac metabolism and angiogenesis during ischemia. J Mol Med (Berl) 2007; 85:1309-15. [PMID: 18026917 DOI: 10.1007/s00109-007-0279-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 10/22/2007] [Accepted: 10/23/2007] [Indexed: 12/12/2022]
Abstract
Myocardial ischemia, the most common cause of cardiac hypoxia in clinical medicine, occurs when oxygen delivery cannot meet myocardial metabolic requirements in the heart. This deficiency can result from either a reduced supply of oxygen (decreased coronary bloodflow) or an increased myocardial demand for oxygen (increased wall stress or afterload). Patients with stable coronary artery disease as well as patients experiencing acute myocardial infarction can experience episodes of severe ischemia. Although hypoxia is an obligatory component, it is not the sole environmental stress experienced by the ischemic heart. Reperfusion after ischemia is associated with increased oxidative stress as the heart reverts to aerobic respiration and thereby generates toxic levels of reactive oxygen species (ROS). During mild ischemia, mitochondrial function is partially compromised and substrate preferences adapt to sustain adequate ATP generation. With severe ischemia, mitochondrial function is markedly compromised and anaerobic metabolism must provide energy no matter what the cost in generation of toxic ROS adducts. Ischemia produces a variety of environmental stresses that impair cardiovascular function. As a result, multiple signaling pathways are activated in mammalian cells during ischemia/reperfusion injury in an attempt to minimize cellular injury and maintain cardiac output. Amongst the transcriptional regulators activated are members of the hypoxia inducible factor (HIF) transcription factor family. HIF factors regulate a variety of genes that affect a myriad of cellular processes including metabolism, angiogenesis, cell survival, and oxygen delivery, all of which are important in the heart. In this review, we will focus on the metabolic and angiogenic aspects of HIF biology as they relate to the heart during ischemia. We will review the metabolic requirements of the heart under normal as well as hypoxic conditions, the effects of preconditioning and its regulation as it pertains to HIF biology, the apparent roles of HIF-1 and HIF-2 in intermediary metabolism, and translational applications of HIF-1 and HIF-2 biology to cardiac angiogenesis. Increased understanding of the role of HIFs in cardiac ischemia will ultimately influence clinical cardiovascular practice.
Collapse
Affiliation(s)
- Ralph V Shohet
- John A. Burns School of Medicine, Center for Cardiovascular Research, University of Hawaii, 651 Ilalo St., Honolulu, HI 96813, USA,
| | | |
Collapse
|