1401
|
Affiliation(s)
- Paolo Rebulla
- Centro Trasfusionale e di Immunologia dei Trapianti, Cell Factory "Franco Calori", IRCCS Ospedale Maggiore, Milano, Italy
| | | |
Collapse
|
1402
|
Dernbach E, Urbich C, Brandes RP, Hofmann WK, Zeiher AM, Dimmeler S. Antioxidative stress-associated genes in circulating progenitor cells: evidence for enhanced resistance against oxidative stress. Blood 2004; 104:3591-7. [PMID: 15161665 DOI: 10.1182/blood-2003-12-4103] [Citation(s) in RCA: 263] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adult and embryonic stem cells hold great promise for regenerative medicine. Expression profiling of stem cells revealed a characteristic imprint of genes, so-called "stemness" genes, providing resistance to stress. Circulating progenitor cells with an endothelial phenotype (EPCs) can be isolated from peripheral blood and contribute to neovascularization and endothelial regeneration. We investigated whether EPCs are equipped with an antioxidative defense to provide resistance against oxidative stress. EPCs exhibited a significantly lower basal reactive oxygen species (ROS) concentration as compared with mature umbilical vein endothelial cells (HUVECs). Incubation with H(2)O(2) (500 microM) or the redox cycler LY-83583 (10 microM) profoundly increased the ROS concentration to 3- and 4-fold and induced apoptosis in HUVECs. In contrast, H(2)O(2) and LY-83583 induced only a minor increase in intracellular ROS levels and apoptosis in EPCs. Consistently, the expression of the intracellular antioxidative enzymes catalase, glutathione peroxidase and manganese superoxide dismutase (MnSOD), was significantly higher in EPCs versus HUVECs and human microvascular endothelial cells. In accordance, combined inhibition of these antioxidative enzymes increased ROS levels in EPCs and impaired EPC survival and migration. Taken together, EPCs reveal a higher expression of antioxidative enzymes and, thus, are exquisitely equipped to be protected against oxidative stress consistent with their progenitor cell character.
Collapse
Affiliation(s)
- Elisabeth Dernbach
- Division of Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
1403
|
Hiasa KI, Ishibashi M, Ohtani K, Inoue S, Zhao Q, Kitamoto S, Sata M, Ichiki T, Takeshita A, Egashira K. Gene transfer of stromal cell-derived factor-1alpha enhances ischemic vasculogenesis and angiogenesis via vascular endothelial growth factor/endothelial nitric oxide synthase-related pathway: next-generation chemokine therapy for therapeutic neovascularization. Circulation 2004; 109:2454-61. [PMID: 15148275 DOI: 10.1161/01.cir.0000128213.96779.61] [Citation(s) in RCA: 227] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stromal cell-derived factor-1alpha (SDF-1alpha) is implicated as a chemokine for endothelial progenitor cells (EPCs). We therefore hypothesized that SDF-1alpha gene transfer would induce therapeutic neovascularization in vivo by functioning as a chemokine of EPC. METHODS AND RESULTS To examine SDF-1alpha-induced mobilization of EPC, we used bone marrow-transplanted mice whose blood cells ubiquitously express beta-galactosidase (LacZ). We produced unilateral hindlimb ischemia in the mice and transfected them with plasmid DNA encoding SDF-1alpha or empty plasmids into the ischemic muscles. SDF-1alpha gene transfer mobilized EPCs into the peripheral blood, augmented recovery of blood perfusion to the ischemic limb, and increased capillary density associated with partial incorporation of LacZ-positive cells into the capillaries of the ischemic limb, suggesting that SDF-1alpha induced vasculogenesis and angiogenesis. SDF-1alpha gene transfer did not affect ischemia-induced expression of vascular endothelial growth factor (VEGF) but did enhance Akt and endothelial nitric oxide synthase (eNOS) activity. Blockade of VEGF or NOS prevented all such SDF-1alpha-induced effects. CONCLUSIONS SDF-1alpha gene transfer enhanced ischemia-induced vasculogenesis and angiogenesis in vivo through a VEGF/eNOS-related pathway. This strategy might become a novel chemokine therapy for next generation therapeutic neovascularization.
Collapse
Affiliation(s)
- Ken-ichi Hiasa
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1404
|
Alessandri G, Emanueli C, Madeddu P. Genetically Engineered Stem Cell Therapy for Tissue Regeneration. Ann N Y Acad Sci 2004; 1015:271-84. [PMID: 15201167 DOI: 10.1196/annals.1302.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Therapeutic angiogenesis/vasculogenesis represents a new approach to treat patients with ischemic disease not curable with conventional treatment. This review focuses on the rationale and preliminary results of combining stem cell and gene therapy for regenerative medicine. Under disease conditions, impaired neovascularization results from diminished vascular growth factor production and primary dysfunction of endothelial cells and their progenitors. Advances in our ability to genetically manipulate cells ex vivo has provided the technological platform to implement stem cell biology and circumvent the potential hazard of direct gene transfer. Ex vivo engineered endothelial progenitor cells have been used for the treatment of peripheral limb ischemia. The approach eliminates the drawback of immune response against viral vectors and makes feasible repeating the therapeutic procedure in case of injury recurrence. The strategy of using stem cells as vectors for curative agents proved to be of value for the treatment of pulmonary hypertension and thrombosis. Transplantation of neural stem cells genetically modified to secrete nerve growth factor was able to ameliorate the death of striatal projection neurons caused by transient focal ischemia in the adult rat. By a similar approach, engineered neural stem cells might be used for treating neurodegenerative disorders. Therefore, genetic manipulation of stem cells opens new avenues for regenerative medicine.
Collapse
Affiliation(s)
- Giulio Alessandri
- Laboratory of Neurobiology and Neuroregenerative Therapy, Carlo Besta Institute, Milan, Italy
| | | | | |
Collapse
|
1405
|
Rabelink TJ, de Boer HC, de Koning EJP, van Zonneveld AJ. Endothelial Progenitor Cells: More Than an Inflammatory Response? Arterioscler Thromb Vasc Biol 2004; 24:834-8. [PMID: 15001453 DOI: 10.1161/01.atv.0000124891.57581.9f] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The formation of new capillaries (angiogenesis) may be of clinical importance in facilitating reperfusion and regeneration of hibernating cardiac tissue after myocardial infarction and in microvascular ischemia. Evidence is accumulating that as part of the response to hypoxia, bone marrow-derived circulating endothelial progenitor cells (CEPs) are mobilized and subsequently differentiate into proper endothelial cells. There are also indications that such CEPs can facilitate endothelial repair and angiogenesis in vivo. It is not clear yet, however, whether these CEPs are essential for these adaptive processes or what the relative contribution of CEP is compared with that of other mononuclear inflammatory cells that are mobilized to areas of ischemia. Moreover, there are still many uncertainties about how cardiovascular risk factors alter CEP function. Particularly when therapeutically mobilizing CEPs, a further understanding of this issue is essential to assess the risk of potentially harmful side effects of altered CEP function.
Collapse
Affiliation(s)
- Ton J Rabelink
- Department of Nephrology, Leiden University Medical Center, The Netherlands.
| | | | | | | |
Collapse
|
1406
|
Kristensen J, Mortensen UM, Nielsen SS, Maeng M, Kaltoft A, Nielsen TT, Rehling M. Myocardial perfusion imaging with 99mTc sestamibi early after reperfusion reliably reflects infarct size reduction by ischaemic preconditioning in an experimental porcine model. Nucl Med Commun 2004; 25:495-500. [PMID: 15100509 DOI: 10.1097/00006231-200405000-00011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Reliable methods for assessment of tissue reperfusion early after revascularizing therapy for acute myocardial infarction are needed. Myocardial perfusion imaging with Tc sestamibi (MIBI MPI) may serve this purpose. Usage during early reperfusion may be problematic e.g. due to ischaemic preconditioning (IP), which is important in inducing ischaemic tolerance. It is mediated through the opening of mitochondrial K ATP channels, reducing mitochondrial membrane potential. This may, as well as ischaemia per se, affect cellular uptake of Tc sestamibi. We therefore studied the reliability of MIBI MPI during early reperfusion as a measure of infarct size and its reduction by ischaemic preconditioning. METHODS AND RESULTS We compared MIBI MPI (cut-off, 45% of maximum pixel count) with a histochemical method in a porcine model, nine controls and eight IP pigs, using 45 min catheter based coronary occlusion of the left anterior descending artery. Infarct size (IS) was determined relative to the area at risk (AAR). The relative infarct size (IS/AAR) after 120 min reperfusion estimated by MPI was 0.83 (0.17) in controls vs 0.07 (0.12) in the IP group (mean (SD), P<0.001). The corresponding values for histochemistry were controls 0.77 (0.19) vs IP 0.07 (0.11), P<0.001. IS/AAR measured by MPI and histochemistry were correlated significantly (r=0.93, P<0.001). Furthermore, IS relative to left ventricular mass (IS/LV) determined by autoradiography and histochemistry correlated (r=0.93, P<0.001). MPI overestimated IS/LV and AAR/LV by approximately a factor of 2 compared with histochemistry or autoradiography. CONCLUSION MIBI MPI during early reperfusion is a reliable measure of relative infarct size reduction after ischaemic preconditioning, supporting use for stratification for adjunctive therapy and for assessment of prognosis.
Collapse
Affiliation(s)
- Jens Kristensen
- Department of Nuclear Medicine, Aarhus University Hospital, Skejby, Brendstrupgaardsvej, DK-8200 Aarhus N, Denmark.
| | | | | | | | | | | | | |
Collapse
|
1407
|
Nygren JM, Jovinge S, Breitbach M, Säwén P, Röll W, Hescheler J, Taneera J, Fleischmann BK, Jacobsen SEW. Bone marrow–derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med 2004; 10:494-501. [PMID: 15107841 DOI: 10.1038/nm1040] [Citation(s) in RCA: 714] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2004] [Accepted: 04/01/2004] [Indexed: 12/11/2022]
Abstract
Recent studies have suggested that bone marrow cells might possess a much broader differentiation potential than previously appreciated. In most cases, the reported efficiency of such plasticity has been rather low and, at least in some instances, is a consequence of cell fusion. After myocardial infarction, however, bone marrow cells have been suggested to extensively regenerate cardiomyocytes through transdifferentiation. Although bone marrow-derived cells are already being used in clinical trials, the exact identity, longevity and fate of these cells in infarcted myocardium have yet to be investigated in detail. Here we use various approaches to induce acute myocardial injury and deliver transgenically marked bone marrow cells to the injured myocardium. We show that unfractionated bone marrow cells and a purified population of hematopoietic stem and progenitor cells efficiently engraft within the infarcted myocardium. Engraftment was transient, however, and hematopoietic in nature. In contrast, bone marrow-derived cardiomyocytes were observed outside the infarcted myocardium at a low frequency and were derived exclusively through cell fusion.
Collapse
Affiliation(s)
- Jens M Nygren
- Hematopoietic Stem Cell Laboratory, Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, BMC B10, Klinikgatan 26, 221 84 Lund, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
1408
|
von Harsdorf R, Poole-Wilson PA, Dietz R. Regenerative capacity of the myocardium: implications for treatment of heart failure. Lancet 2004; 363:1306-13. [PMID: 15094278 DOI: 10.1016/s0140-6736(04)16006-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Research into myocardial regeneration has an exciting future, shown by the results of experimental and clinical work challenging the dogma that the heart is a postmitotic non-regenerating organ. Such studies have initiated a lively debate about the feasibility of novel treatment approaches leading to the recovery of damaged myocardial tissue. The possibility of reconstituting dead myocardium by endogenous cardiomyocyte replication, transplantation, or activation of stem cells--or even cloning of an artificial heart--is being advanced, and will be a major subject of future research. Although health expenditure for heart failure in the industrial world is high, we are still a long way from being able to treat the cause of reduced myocardial contractility. Despite the hopes of some people, conventional treatment for heart failure does not achieve myocardial regeneration. We present a virtual case report of a patient with acute myocardial infarction; we discuss treatment options, including strategies aimed at organ regeneration.
Collapse
Affiliation(s)
- Rüdiger von Harsdorf
- Department of Cardiology, Campus Virchow Clinic, Charité, Humboldt University Berlin, Berlin, Germany.
| | | | | |
Collapse
|
1409
|
Verma S, Kuliszewski MA, Li SH, Szmitko PE, Zucco L, Wang CH, Badiwala MV, Mickle DAG, Weisel RD, Fedak PWM, Stewart DJ, Kutryk MJB. C-reactive protein attenuates endothelial progenitor cell survival, differentiation, and function: further evidence of a mechanistic link between C-reactive protein and cardiovascular disease. Circulation 2004; 109:2058-67. [PMID: 15078802 DOI: 10.1161/01.cir.0000127577.63323.24] [Citation(s) in RCA: 401] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Myocardial ischemia provides a potent stimulus to angiogenesis, and the mobilization and differentiation of endothelial progenitor cells (EPCs) has been shown to be important in this process. An elevated level of C-reactive protein (CRP) has emerged as one of the most powerful predictors of cardiovascular disease. However, the impact of CRP on EPC biology is unknown. METHODS AND RESULTS EPCs were isolated from the peripheral venous blood of healthy male volunteers. Cells were cultured in endothelial cell basal medium-2 in the absence and presence of CRP (5 to 20 microg/mL), rosiglitazone (1 micromol/L), and/or vascular endothelial growth factor. EPC differentiation, survival, and function were assayed. CRP at concentrations > or =15 microg/mL significantly reduced EPC cell number, inhibited the expression of the endothelial cell-specific markers Tie-2, EC-lectin, and VE-cadherin, significantly increased EPC apoptosis, and impaired EPC-induced angiogenesis. EPC-induced angiogenesis was dependent on the presence of nitric oxide, and CRP treatment caused a decrease in endothelial nitric oxide synthase mRNA expression by EPCs. However, all of these detrimental CRP-mediated effects on EPCs were attenuated by pretreatment with rosiglitazone, a peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist. CONCLUSIONS Human recombinant CRP, at concentrations known to predict adverse vascular outcomes, directly inhibits EPC differentiation, survival, and function, key components of angiogenesis and the response to chronic ischemia. This occurs in part via an effect of CRP to reduce EPC eNOS expression. The PPARgamma agonist rosiglitazone inhibits the negative effects of CRP on EPC biology. The ability of CRP to inhibit EPC differentiation and survival may represent an important mechanism that further links inflammation to cardiovascular disease.
Collapse
Affiliation(s)
- Subodh Verma
- Division of Cardiac Surgery, Toronto General Hospital, 14EN-215, 200 Elizabeth St, Toronto, Ontario, Canada M5G 2C4.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1410
|
Abstract
Atherosclerotic vascular disease becomes a clinical problem when there is sufficient atherosclerotic plaque burden and/or endothelial dysfunction to cause a limitation of nutrient blood flow to tissues. However, once myocardial infarction has occurred, there is little, if any, way to stimulate the growth of new blood vessels or cardiac muscle to replace that which has been lost. The potential use of hematopoietic stem cells (HSCs) to treat cardiovascular disease has recently been suggested from preclinical and clinical studies. HSCs are precursors of all the blood cells, but they may also give rise to cells of the vascular system, endothelial cells in the form of endothelial progenitor cells (EPCs). Clinical trials have been conducted in patients with either acute myocardial infarction or limb ischemia to determine the initial effectiveness and safety of this treatment approach. These studies demonstrated the potential clinical effectiveness of this stem cell approach to the treatment of patients with acute myocardial ischemia and limb ischemia. Today, more preclinical studies are planned to elucidate the mechanism by which transplanted stem cells can home and differentiate into these endothelial cells and cardiac muscle cells. At the same time, new clinical trials are planned to evaluate both chronic, stable as well as acute myocardial ischemia and limb ischemia with CD34+ and CD133+ stem cells, as well as with further selected EPCs and mesenchymal stem cells.
Collapse
Affiliation(s)
- David L Amrani
- Baxter Healthcare, RLT-12 Route 120 and Wilson Rd, Round Lake, IL 60073, USA.
| | | |
Collapse
|
1411
|
Li TS, Hayashi M, Liu ZL, Ito H, Mikamo A, Furutani A, Matsuzaki M, Hamano K. Low angiogenic potency induced by the implantation of ex vivo expanded CD117+stem cells. Am J Physiol Heart Circ Physiol 2004; 286:H1236-41. [PMID: 14656709 DOI: 10.1152/ajpheart.00950.2003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ex vivo expansion of stem cells might be a feasible method of resolving the problem of limited cell supply in cell-based therapy. The implantation of expanded CD34+endothelial progenitor cells has the capacity to induce angiogenesis. In this study, we tried to induce angiogenesis by implanting expanded CD117+stem cells derived from mouse bone marrow. After 2 wk of culture with the addition of several growth factors, the CD117+stem cells expanded ∼20-fold and had an endothelial phenotype with high expression of CD34 and vascular endothelial-cadherin. However, >70% of these ex vivo expanded cells had a senescent phenotype by β-galactosidase staining, and their survival and incorporation were poor after implantation into the ischemic limbs of mice. Compared with the PBS injection only, the microvessel density and the percentage of limb blood flow were significantly higher after the implantation of 2 × 105freshly collected CD117+cells ( P < 0.01) but not after the implantation of 2 × 105expanded CD117+cells ( P > 0.05). These data indicate that ex vivo expansion of CD117+stem cells has low potency for inducing therapeutic angiogenesis, which might be related to the cellular senescence during ex vivo expansion.
Collapse
Affiliation(s)
- Tao-Sheng Li
- Department of Medical Bioregulation, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | |
Collapse
|
1412
|
Abstract
Cardiovascular disease is a global cause of mortality and morbidity. Current treatments fail to address the underlying scarring and cell loss, which are the causes of ischaemic heart failure. Cellular transplantation can overcome these problems and new impetus has been injected into this field following the isolation of human embryonic and adult stem cells. These cells have shown remarkable ability to produce cardiomyocytes and vascular cells in vitro and in vivo. Initial transplantation studies have demonstrated functional benefits and it is hoped further randomised clinical trials will concur with initial findings. Much basic science remains to be unearthed, such as the signals for homing, differentiation and engraftment of transplanted cells. Further matters of concern are the role of cell fusion and the mechanisms by which transplanted cells improve cardiac function. In spite of initial progress made in stem cell therapy there is still much to be done and we are some way off from achieving the goal of effective cellular regeneration.
Collapse
Affiliation(s)
- M J Lovell
- Department of Clinical Pharmacology, William Harvey Research Institute, Bart's and the London, London, UK.
| | | |
Collapse
|
1413
|
Beller GA. First annual Mario S. Verani, MD, Memorial lecture: clinical value of myocardial perfusion imaging in coronary artery disease. J Nucl Cardiol 2004; 10:529-42. [PMID: 14569247 DOI: 10.1016/s1071-3581(03)00655-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
I am honored to give this lecture in memory of Dr Mario S. Verani, a friend and a respected colleague in the field of nuclear cardiology. Dr Verani made many immense contributions to the field of nuclear cardiology during his lifetime. He was a pioneer in the field of pharmacologic stress imaging and played a leading role in validating the utility of intravenous adenosine infusion for stress imaging in detecting significant coronary stenoses and assessing prognosis. He and his colleagues at Baylor reported that pharmacologic stress imaging could separate high- and low-risk patient subsets after acute myocardial infarction, particularly among patients who underwent thrombolytic therapy. He informed us that certain drugs that patients took could influence perfusion defect severity. This work led to a clinical study showing that ischemic defect size could be substantially reduced with medical therapy in patients who had recently had an acute infarction. An outcome of this observation was the launching of the INSPIRE (adenosINe technetium-99m Sestamibi single-photon emission computed tomography Post-InfaRction Evaluation) trial in which postinfarction patients are randomized to medical therapy or revascularization. Dr Verani was an able debater when assigned the position for advocating for stress radionuclide perfusion imaging against stress echocardiography. He engaged in these debates with substantial knowledge but always with a touch of humor. Dr Verani's courage was an inspiration to all of us during his illness. His legacy as a pioneer will endure as the field of nuclear cardiology continues to mature and expand.
Collapse
Affiliation(s)
- George A Beller
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
1414
|
Reinlib L, Abraham W. Recovery from heart failure with circulatory assist: a working group of the National, Heart, Lung, and Blood Institute. J Card Fail 2004; 9:459-63. [PMID: 14966786 DOI: 10.1016/s1071-9164(03)00592-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Over the past decade, mechanical circulatory support has been beneficial as a bridge to cardiac transplantation, and anecdotal evidence suggests that heart failure patients fitted with mechanical assist devices experience direct cardiac benefits. Moreover, recent trials on limited numbers and subpopulations of patients--notably the Randomized Evaluation of Mechanical Assistance for the Treatment of Congestive Heart Failure (REMATCH)--support earlier observations of improved cardiac function and point towards the use of assist devices as destination therapy. Methods and results To investigate this phenomenon, on August 2-3, 2001, the National Heart, Lung, and Blood Institute convened the working group, "Recovery from Heart Failure with Circulatory Assist" in Bethesda, Maryland. The team included cardiac surgeons, cardiologists, and experts in experimental research. The goal was to prioritize recommendations to guide future programs in: (1). elucidating the mechanisms leading to reverse remodeling associated with a left ventricular assist device (LVAD); (2). exploring advanced treatments, including novel pharmacologies, tissue engineering, and cell therapies, to optimize recovery with LVAD therapy; and (3). identifying target genes, proteins, and cellular pathways to focus on for production of novel therapies for myocardial recovery and cardiovascular disease. CONCLUSIONS The working group also made research and clinical recommendations to eventually translate findings into improved therapeutic strategies and device design: (1). support collaborations among clinical and basic scientists with an emphasis on clinical/translational research that might eventually lead to clinical trials; (2). identify candidate patients most likely to benefit from LVAD as a destination therapy; (3). explore potential biomarkers indicating when patients could most successfully be weaned from devices; and (4). promote clinical and experimental study of mechanically assisted organs and the tissue derived from them.
Collapse
Affiliation(s)
- Leslie Reinlib
- Division of Heart and Vascular Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, USA
| | | |
Collapse
|
1415
|
Heeschen C, Lehmann R, Honold J, Assmus B, Aicher A, Walter DH, Martin H, Zeiher AM, Dimmeler S. Profoundly reduced neovascularization capacity of bone marrow mononuclear cells derived from patients with chronic ischemic heart disease. Circulation 2004; 109:1615-22. [PMID: 15037527 DOI: 10.1161/01.cir.0000124476.32871.e3] [Citation(s) in RCA: 467] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cell therapy with bone marrow-derived stem/progenitor cells is a novel option for improving neovascularization and cardiac function in ischemic heart disease. Circulating endothelial progenitor cells in patients with coronary heart disease are impaired with respect to number and functional activity. However, whether this impairment also extends to bone marrow-derived mononuclear cells (BM-MNCs) in patients with chronic ischemic cardiomyopathy (ICMP) is unclear. METHODS AND RESULTS BM-MNCs were isolated from bone marrow aspirates in 18 patients with ICMP (ejection fraction, 38+/-11%) and 8 healthy control subjects (controls). The number of hematopoietic stem/progenitor cells (CD34+/CD133+), CD49d(+) (VLA-4) cells, and CXCR4+ cells did not differ between the 2 groups. However, the colony-forming capacity of BM-MNCs from patients with ICMP was significantly lower compared with BM-MNCs from healthy controls (37.3+/-25.0 versus 113.8+/-70.4 granulocyte-macrophage colony-forming units; P=0.009). Likewise, the migratory response to stromal cell-derived factor 1 (SDF-1) and vascular endothelial growth factor (VEGF) was significantly reduced in BM-MNCs derived from patients with ICMP compared with BM-MNCs from healthy controls (SDF-1, 46.3+/-26.2 versus 108.6+/-40.4 cells/microscopic field, P<0.001; VEGF, 34+/-24.2 versus 54.8+/-29.3 cells/microscopic field, P=0.027). To assess the in vivo relevance of these findings, we tested the functional activity of BM-MNCs to improve neovascularization in a hindlimb animal model using nude mice. Two weeks after ligation of the femoral artery and intravenous injection of 5x10(5) BM-MNCs, laser Doppler-derived relative limb blood flow in mice treated with BM-MNCs from patients with ICMP was significantly lower compared with mice treated with BM-MNCs from healthy controls (0.45+/-0.14 versus 0.68+/-0.15; P<0.001). The in vivo neovascularization capacity of BM-MNCs closely correlated with the in vitro assessment of SDF-1-induced migration (r=0.78; P<0.001) and colony-forming capacity (r=0.74; P<0.001). CONCLUSIONS BM-MNCs isolated from patients with ICMP have a significantly reduced migratory and colony-forming activity in vitro and a reduced neovascularization capacity in vivo despite similar content of hematopoietic stem cells. This functional impairment of BM-MNCs from patients with ICMP may limit their therapeutic potential for clinical cell therapy.
Collapse
Affiliation(s)
- Christopher Heeschen
- Molecular Cardiology, Department of Internal Medicine IV, University of Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
1416
|
Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KBS, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 2004; 428:664-8. [PMID: 15034593 DOI: 10.1038/nature02446] [Citation(s) in RCA: 1477] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2003] [Accepted: 03/01/2004] [Indexed: 12/21/2022]
Abstract
The mammalian heart has a very limited regenerative capacity and, hence, heals by scar formation. Recent reports suggest that haematopoietic stem cells can transdifferentiate into unexpected phenotypes such as skeletal muscle, hepatocytes, epithelial cells, neurons, endothelial cells and cardiomyocytes, in response to tissue injury or placement in a new environment. Furthermore, transplanted human hearts contain myocytes derived from extra-cardiac progenitor cells, which may have originated from bone marrow. Although most studies suggest that transdifferentiation is extremely rare under physiological conditions, extensive regeneration of myocardial infarcts was reported recently after direct stem cell injection, prompting several clinical trials. Here, we used both cardiomyocyte-restricted and ubiquitously expressed reporter transgenes to track the fate of haematopoietic stem cells after 145 transplants into normal and injured adult mouse hearts. No transdifferentiation into cardiomyocytes was detectable when using these genetic techniques to follow cell fate, and stem-cell-engrafted hearts showed no overt increase in cardiomyocytes compared to sham-engrafted hearts. These results indicate that haematopoietic stem cells do not readily acquire a cardiac phenotype, and raise a cautionary note for clinical studies of infarct repair.
Collapse
Affiliation(s)
- Charles E Murry
- Department of Pathology, Box 357470, Room D-514 HSB, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1417
|
Xaymardan M, Tang L, Zagreda L, Pallante B, Zheng J, Chazen JL, Chin A, Duignan I, Nahirney P, Rafii S, Mikawa T, Edelberg JM. Platelet-Derived Growth Factor-AB Promotes the Generation of Adult Bone Marrow–Derived Cardiac Myocytes. Circ Res 2004; 94:E39-45. [PMID: 14963008 DOI: 10.1161/01.res.0000122042.51161.b6] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The directed generation of cardiac myocytes from endogenous stem cells offers the potential for novel therapies for cardiovascular disease. To facilitate the development of such approaches, we sought to identify and exploit the pathways directing the generation of cardiac myocytes from adult rodent bone marrow cells (BMCs). In vitro cultures supporting the spontaneous generation of functional cardiac myocytes from murine BMCs demonstrated induced expression of platelet-derived growth factor (PDGF)-A and -B isoforms with α- and β-myosin heavy chains as well as connexin43. Supplementation of PDGF-AB speeded the kinetics of myocyte development in culture by 2-fold. In a rat heart, myocardial infarction pretreatment model PDGF-AB also promoted the derivation of cardiac myocytes from BMCs, resulting in a significantly greater number of islands of cardiac myocyte bundles within the myocardial infarction scar compared with other treatment groups. However, gap junctions were detected only between the cardiac myocytes receiving BMCs alone, but not BMCs injected with PDGF-AB. Echocardiography and exercise testing revealed that the functional improvement of hearts treated with the combination of BMCs and PDGF-AB was no greater than with injections of BMCs or PDGF-AB alone. These studies demonstrated that PDGF-AB enhances the generation of BMC-derived cardiac myocytes in rodent hearts, but suggest that alterations in cellular patterning may limit the functional benefit from the combined injection of PDGF-AB and BMCs. Strategies based on the synergistic interactions of PDGF-AB and endogenous stem cells will need to maintain cellular patterning in order to promote the restoration of cardiac function after acute coronary occlusion. The full text of this article is available online at http://circres.ahajournals.org.
Collapse
Affiliation(s)
- Munira Xaymardan
- Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1418
|
Affiliation(s)
- Hiroaki Matsubara
- Department of Cardiovascular Medicine, Kyoto Prefectural University School of Medicine, Translational Research Center, Kyoto University Hospital, Kyoto, Japan.
| |
Collapse
|
1419
|
Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ, Soo Lee D, Sohn DW, Han KS, Oh BH, Lee MM, Park YB. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. Lancet 2004; 363:751-6. [PMID: 15016484 DOI: 10.1016/s0140-6736(04)15689-4] [Citation(s) in RCA: 548] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bone-marrow stem-cell transplantation has been shown to improve cardiac function in patients with myocardial infarction. We examined the feasibility and efficacy of granulocyte-colony stimulating factor (G-CSF) therapy and subsequent intracoronary infusion of collected peripheral blood stem-cells (PBSCs) in such patients. METHODS We prospectively randomised 27 patients with myocardial infarction who underwent coronary stenting for the culprit lesion of infarction into three groups; cell infusion (n=10), G-CSF alone (n=10), and control group (n=7). Changes in left ventricular systolic function and perfusion were assessed after 6 months. By December, 2003, seven patients from the cell infusion group, three from the G-CSF group, and one from the control group had been assessed. FINDINGS G-CSF injection and intracoronary infusion of the mobilised PBSC did not aggravate inflammation and ischaemia during the periprocedural period. Exercise capacity (mean treadmill exercise time: 450 s [SD 178] at baseline vs 578 s [168] at 6 months' follow-up, p=0.004), myocardial perfusion (perfusion defect 11.6% [9.6] vs 5.3% [5.0], p=0.020) and systolic function (left ventricular ejection fraction 48.7% [8.3] vs 55.1% [7.4], p=0.005) improved significantly in patients who received cell infusion. However, we noted an unexpectedly high rate of in-stent restenosis at culprit lesion in patients who received G-CSF, and therefore we stopped enrollment. INTERPRETATION G-CSF therapy with intracoronary infusion of PBSC showed improved cardiac function, and promoted angiogenesis in patients with myocardial infarction. However, aggravation of restenosis could be a serious problem. In future studies with G-CSF based stem-cell therapy, patients should be carefully monitored for unexpected effects.
Collapse
Affiliation(s)
- Hyun-Jae Kang
- Cardiovascular Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1420
|
Abstract
Molecular imaging is a rapidly evolving discipline with the goal of developing tools to display and quantify molecular and cellular targets in vivo. The heart of this field is based on the rational design and screening of targeted and activatable imaging reporter agents to sense fundamental processes of biology. Parallel advances in small animal imaging systems and in agent synthesis have allowed molecular imaging applications to extend into the in vivo arena. These advances have permitted, for example, in vivo sensing of inflammation, apoptosis, cell trafficking, and gene expression. In this review, we first review core principles of molecular imaging with an emphasis on smart, activatable agent technology. We then discuss applications of state-of-the-art molecular probes to interrogate important aspects of cardiovascular biology, with a focus on atherosclerosis, thrombosis, and heart failure. In the ensuing years, we anticipate that fundamental aspects of cardiovascular biology will be detectable in vivo, and that promising molecular imaging agents will be translated into the clinical arena to guide diagnosis and therapy of human cardiovascular illness.
Collapse
Affiliation(s)
- Farouc A Jaffer
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Mass 02129, USA.
| | | |
Collapse
|
1421
|
Abstract
PURPOSE OF REVIEW The last few years have witnessed a growing interest in regenerative therapy of the failing heart by cell transplantation. Special emphasis has been put on skeletal myoblasts and bone marrow-derived stem cells, with a flurry of experimental studies generating overall positive but occasionally conflicting results. It is thus appropriate to review the most important of these studies in light of the major issues that still impede widespread clinical use of cell therapy. RECENT FINDINGS Recent laboratory data demonstrate the ability of autologous skeletal myoblasts to engraft into scarred myocardium and improve its function. Equally successful results have been reported with bone marrow-derived cells which, in contrast to myoblasts, are credited with a plasticity that might allow their transdifferentiation into cardiac or endothelial cells in response to organ-specific cues. However, some major questions remain unanswered; they include the choice of the optimal cell type in relation with the target patient population, the strategies for enhancing cell survival and functional integration, the clarification of the mechanisms of improvement, and the means of reducing invasiveness of cell delivery. SUMMARY Although laboratory research attempts to overcome these persisting hurdles, the accumulated body of evidence warrants implementation of clinical trials. The earliest ones have now documented the feasibility of cell therapy. It is now appropriate to conduct safety and efficacy studies which, if carefully done, should allow assessment of the extent to which this concept of regenerative therapy can be made a clinical reality.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery & INSERM U 572, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
1422
|
Chachques JC, Acar C, Herreros J, Trainini JC, Prosper F, D'Attellis N, Fabiani JN, Carpentier AF. Cellular cardiomyoplasty: clinical application. Ann Thorac Surg 2004; 77:1121-30. [PMID: 14992951 DOI: 10.1016/j.athoracsur.2003.09.081] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Myocardial regeneration can be induced with the implantation of a variety of myogenic and angiogenic cell types. More than 150 patients have been treated with cellular cardiomyoplasty worldwide, 18 patients have been treated by our group. Cellular cardiomyoplasty seems to reduce the size and fibrosis of infarct scars, limit postischemic remodelling, and restore regional myocardial contractility. Techniques for skeletal myoblasts culture and ex vivo expansion using autologous patient serum (obtained from plasmapheresis) have been developed by our group. In this article we propose (1) a total autologous cell culture technique and procedures for cell delivery and (2) a clinical trial with appropriate endpoints structured to determine the efficacy of cellular cardiomyoplasty.
Collapse
Affiliation(s)
- Juan C Chachques
- Department of Cardiovascular Surgery, European Hospital Georges Pompidou, University of Paris, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
1423
|
|
1424
|
Lee S, Bick-Forrester J, Makkar RR, Forrester JS. Stem-Cell Repair of Infarcted Myocardium: Ready for Clinical Application? ACTA ACUST UNITED AC 2004; 2:100-6. [PMID: 15604853 DOI: 10.1111/j.1541-9215.2004.03436.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In cell culture, adult bone marrow stem cells can develop the phenotypic characteristics of myocytes and endothelial cells, and express myocyte-specific and endothelium-specific proteins. In subsequent animal laboratory studies and early clinical trials, stem cells have been delivered to infarcted myocardium by direct injection, by intravascular injection, and by bone marrow stimulation. In animals, myocyte apoptosis is reduced, capillary density increases, and regional perfusion increases accompanied by a decrease in infarct size. Early clinical trials indicate that a variety of approaches is technically feasible and safe in the short term. The trials suggest that stem-cell therapy may induce a modest preservation of cardiac function. The methodology for clinical application of stem-cell therapy currently exists in most large heart hospitals. Nonetheless, until theoretical and practical limitations are resolved, it seems prudent to confine this therapy to randomized clinical trials.
Collapse
Affiliation(s)
- Steve Lee
- Division of Cardiology, Cedars-Sinai Medical Center, University of California Los Angeles School of Medicine, Los Angeles, CA 90048, USA
| | | | | | | |
Collapse
|
1425
|
Liddell RP, Weiss CR, Hofmann LV. Therapeutic angiogenesis: the next frontier for interventional radiology. Tech Vasc Interv Radiol 2004; 7:40-8. [PMID: 15071780 DOI: 10.1053/j.tvir.2004.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The field of interventional radiology has traditionally relied on mechanical methods to treat vascular disease, such as angioplasty balloons and stents. Although there have been a number of important technical advances in endovascular devices, there are still a number of patients who are not candidates for percutaneous or surgical revascularization. As we approach the technical limits of these newer devices, therapeutic angiogenesis may play an ever-increasing role in the future. Interventional radiologists have unique delivery skills that would complement the on-going research in this area. It is the goal of this article to serve as a primer for interventional radiologists on the agents and techniques used in this exciting field.
Collapse
Affiliation(s)
- Robert P Liddell
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | |
Collapse
|
1426
|
Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation 2004; 109:1292-8. [PMID: 14993122 DOI: 10.1161/01.cir.0000121425.42966.f1] [Citation(s) in RCA: 1695] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The delivery of autologous cells to increase angiogenesis is emerging as a treatment option for patients with cardiovascular disease but may be limited by the accessibility of sufficient cell numbers. The beneficial effects of delivered cells appear to be related to their pluripotency and ability to secrete growth factors. We examined nonadipocyte stromal cells from human subcutaneous fat as a novel source of therapeutic cells. METHODS AND RESULTS Adipose stromal cells (ASCs) were isolated from human subcutaneous adipose tissue and characterized by flow cytometry. ASCs secreted 1203+/-254 pg of vascular endothelial growth factor (VEGF) per 10(6) cells, 12 280+/-2944 pg of hepatocyte growth factor per 10(6) cells, and 1247+/-346 pg of transforming growth factor-beta per 10(6) cells. When ASCs were cultured in hypoxic conditions, VEGF secretion increased 5-fold to 5980+/-1066 pg/10(6) cells (P=0.0016). The secretion of VEGF could also be augmented 200-fold by transfection of ASCs with a plasmid encoding VEGF (P<0.05). Conditioned media obtained from hypoxic ASCs significantly increased endothelial cell growth (P<0.001) and reduced endothelial cell apoptosis (P<0.05). Nude mice with ischemic hindlimbs demonstrated marked perfusion improvement when treated with human ASCs (P<0.05). CONCLUSIONS Our experiments delineate the angiogenic and antiapoptotic potential of easily accessible subcutaneous adipose stromal cells by demonstrating the secretion of multiple potentially synergistic proangiogenic growth factors. These findings suggest that autologous delivery of either native or transduced subcutaneous ASCs, which are regulated by hypoxia, may be a novel therapeutic option to enhance angiogenesis or achieve cardiovascular protection.
Collapse
Affiliation(s)
- Jalees Rehman
- Krannert Institute of Cardiology, Indiana Center for Vascular Biology & Medicine, Indianapolis, Ind 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1427
|
Tomoda H, Aoki N. Coronary blood flow in evolving myocardial infarction preceded by preinfarction angina: a critical reevaluation of preconditioning effects in clinical cases. Angiology 2004; 55:9-15. [PMID: 14759084 DOI: 10.1177/000331970405500102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study was undertaken to reevaluate the protective effects of preinfarction (pre-MI) angina in acute MI. The mechanisms involved in the apparent protective effects of pre-MI angina have been presumed to be preconditioning effects as defined by experimental studies. The phenomenon, has not, however, been observed in diabetic and/or elderly patients or in those treated by primary percutaneous coronary intervention (PCI). A total of 202 patients with anterior wall MI without a history of MI who underwent primary PCI with coronary balloon dilation and stenting (rate: 50%) <6 hours after onset were studied. Patients included 59 with pre-MI angina (group 1) and 143 without pre-MI angina (group 2). The infarct-related coronary artery was patent on admission in 46% of group 1 and 31% of group 2 (p=0.045). Thrombolysis in Myocardial Infarction (TIMI) 1-2 flow was significantly more frequent in group 1 (29%) than in group 2 (11%, p=0.005) on admission. Among risk factors, clinical background, coronary anatomy, and clinical outcome, the only significant predictor of pre-MI angina was a patent infarct-related coronary artery on admission (odds ratio: 2.39, p = 0.015). There was no significant difference in left ventricular ejection fraction, peak creatine kinase, or the incidences of heart failure and in-hospital/follow-up deaths between these groups. In conclusion, the findings suggest that the protective effects reported in MI with pre-MI angina treated by thrombolysis are due to more fragile thrombotic occlusion, which can be more easily recanalized by thrombolysis, whereas the beneficial effects are not evident in those treated by primary PCI.
Collapse
Affiliation(s)
- Haruo Tomoda
- Department of Cardiology, Tokai University Hospital, Isehara, Kanagawa, Japan.
| | | |
Collapse
|
1428
|
Adams V, Lenk K, Linke A, Lenz D, Erbs S, Sandri M, Tarnok A, Gielen S, Emmrich F, Schuler G, Hambrecht R. Increase of circulating endothelial progenitor cells in patients with coronary artery disease after exercise-induced ischemia. Arterioscler Thromb Vasc Biol 2004; 24:684-90. [PMID: 14988094 DOI: 10.1161/01.atv.0000124104.23702.a0] [Citation(s) in RCA: 244] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The concept of neovascularization in response to tissue ischemia has been extended by the finding of postnatal vasculogenesis initiated by endothelial progenitor cells (EPCs). The aim of this study was to analyze whether a maximal stress test in patients with coronary artery disease (CAD) increases the number of circulating EPCs. METHODS AND RESULTS Blood concentration of EPCs was analyzed by FACS and cell culture assay in CAD patients with (n=16) or without (n=12) exercise-induced myocardial ischemia and in healthy subjects (n=11) for up to 144 hours after maximal stress test. Plasma concentrations of vascular endothelial growth factor (VEGF), basic fibroblast growth factor, tumor necrosis factor-alpha, and granulocyte macrophage-colony stimulating factor were determined by ELISA. EPCs increased significantly in ischemic patients, with a maximum after 24 to 48 hours (cell culture: 3.3+/-0.5-fold increase; FACS: 3.1+/-0.6-fold increase) and returned to baseline within 72 hour. In nonischemic patients and healthy subjects, no EPC increase was detectable. VEGF levels in ischemic patients increased significantly after 2 to 6 hours (maximum after 2 hours; 4.0+/-1.1-fold increase) and no change was observed in nonischemic patients and healthy subjects; DeltaVEGF and DeltaEPC correlated significantly (r=0.66). CONCLUSIONS Patients with symptomatic CAD respond to a single episode of exercise-induced myocardial ischemia with a time-dependent increase in circulating EPCs. This increase may be related to and preceded by an increase in plasma VEGF.
Collapse
Affiliation(s)
- Volker Adams
- University Leipzig, Heart Center Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1429
|
Loomans CJM, Dao HH, van Zonneveld AJ, Rabelink TJ. Is endothelial progenitor cell dysfunction involved in altered angiogenic processes in patients with hypertension? Curr Hypertens Rep 2004; 6:51-4. [PMID: 14972094 DOI: 10.1007/s11906-004-0011-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypertension represents one of the most frequent modifiable risk factors for cardiovascular disease (CVD). Despite the arrival of novel antihypertensive drugs and progress in primary prevention and screening, the improvement of blood pressure control does not similarly reduce the incidence of end-organ damage associated with hypertension. Recently, the concept of reduced angiogenesis as a new CVD risk factor has gained the interest of an increasing number of investigators in the field. Indeed, pharmacologic and stem cell-based strategies aimed to induce angiogenesis seem to improve the course of ischemic vascular disease. However, few to no reports have studied the possible therapeutic potential of angiogenesis in essential hypertension. In this review, we discuss the potential involvement of endothelial progenitor cells in modulating angiogenesis and their putative therapeutic role in improving capillary rarefaction and arterial stiffness in essential hypertensive patients.
Collapse
Affiliation(s)
- Cindy J M Loomans
- Department of Vascular Medicine, University Medical Center of Utrecht, G02.228, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | | | | | | |
Collapse
|
1430
|
Smiseth OA, Steg PG, Sipido K, Battler A. News from the European Society of Cardiology Congress in Vienna, August 30 to September 3, 2003. J Am Coll Cardiol 2004; 43:691-7. [PMID: 14975484 DOI: 10.1016/j.jacc.2003.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Accepted: 11/25/2003] [Indexed: 11/18/2022]
Affiliation(s)
- Otto A Smiseth
- Department of Cardiology, Rikshospitalet University Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
1431
|
Abstract
PURPOSE OF REVIEW Gene therapy utilizes viral and non-viral vectors to transfer genetic material into a host in the hope of treating disease. This article will review the potential applications of gene therapy in the treatment of cardiac and pulmonary diseases. RECENT FINDINGS The results from several phase I and II clinical trials have recently been published. In patients with ischemic heart disease, evidence of coronary revascularization has been observed after the delivery of angiogenic factors. Several trials have demonstrated a reduction in anginal symptoms, increases in exercise tolerance, and objective improvements in myocardial perfusion. Evidence of the transfer of therapeutic genes has been observed in human trials of inherited pulmonary diseases. Unfortunately, there has been little evidence of clinical efficacy in these studies. A variety of gene therapy strategies are being explored in the treatment of thoracic malignancies. Partial antitumor responses have occurred in some of the subjects enrolled in these studies. SUMMARY Significant progress has been made in the field of gene therapy in the past decade. Data from these early animal and human clinical trials will provide important information to guide future studies.
Collapse
Affiliation(s)
- Joseph W Szokol
- Department of Anesthesiology, Evanston Northwestern Healthcare, Evanston and Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60201, USA.
| | | | | | | |
Collapse
|
1432
|
Cleland JGF, Freemantle N, Kaye G, Nasir M, Velavan P, Lalukota K, Mudawi T, Shelton R, Clark AL, Coletta AP. Clinical trials update from the American Heart Association meeting: Ω-3 fatty acids and arrhythmia risk in patients with an implantable defibrillator, ACTIV in CHF, VALIANT, the Hanover autologous bone marrow transplantation study, SPORTIF V, ORBIT and PA. Eur J Heart Fail 2004; 6:109-15. [PMID: 15012926 DOI: 10.1016/j.ejheart.2003.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2003] [Accepted: 12/05/2003] [Indexed: 10/26/2022] Open
Abstract
The American Heart Association meeting reported the results of several clinical trials of particular interest to those who care for patients with heart failure. Omega-3 fatty acids were associated with a trend to increased recurrence of ventricular arrhythmias but not mortality in patients with an implantable debrillator. The ACTIV in CHF study provides more evidence of a therapeutic role for arginine vasopressin antagonists in the treatment of heart failure. The VALIANT study provides further evidence to suggest that a combination of angiotensin receptor antagonist and ACE inhibitor does not reduce mortality but may reduce morbidity in post-MI patients with heart failure or major LV systolic dysfunction. A study of autologous bone marrow cell transplantation into myocardial scar give gave encouraging results. SPORTIF V showed ximelagation to be as effective as warfarin but with improved safety. ORBIT and PAD showed public access defibrillators saved lives but questioned their cost effectiveness. DEFINITE supported a role for ICDs in patients with non-ischemic cardiomyopathy, although cost-effectiveness remains in doubt.
Collapse
Affiliation(s)
- John G F Cleland
- Department of Cardiology, University of Hull, Castle Hill Hospital, Cottingham, Kingston-upon-Hull, HU15 5JQ, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1433
|
Planat-Benard V, Silvestre JS, Cousin B, André M, Nibbelink M, Tamarat R, Clergue M, Manneville C, Saillan-Barreau C, Duriez M, Tedgui A, Levy B, Pénicaud L, Casteilla L. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation 2004; 109:656-63. [PMID: 14734516 DOI: 10.1161/01.cir.0000114522.38265.61] [Citation(s) in RCA: 1262] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Adipose tissue development and remodeling are closely associated with the growth of vascular network. We hypothesized that adipose tissue may contain progenitor cells with angiogenic potential and that therapy based on adipose tissue-derived progenitor cells administration may constitute a promising cell therapy in patients with ischemic disease. METHODS AND RESULTS In mice, cultured stromal-vascular fraction (SVF) cells from adipose tissue have a great proangiogenic potential, comparable to that of bone marrow mononuclear cells in the mouse ischemic hindlimb model. Similarly, cultured human SVF cells differentiate into endothelial cells, incorporate into vessels, and promote both postischemic neovascularization in nude mice and vessel-like structure formation in Matrigel plug. In vitro, these cells represent a homogeneous population of CD34- and CD13-positive cells, which can spontaneously express the endothelial cell markers CD31 and von Willebrand factor when cultured in semisolid medium. Interestingly, dedifferentiated mature human adipocytes have the potential to rapidly acquire the endothelial phenotype in vitro and to promote neovascularization in ischemic tissue and vessel-like structure formation in Matrigel plug, suggesting that cells of endothelial and adipocyte phenotypes may have a common precursor. CONCLUSIONS This study demonstrates, for the first time, that adipocytes and endothelial cells have a common progenitor. Such adipose lineage cells participate in vascular-like structure formation in Matrigel plug and enhance the neovascularization reaction in ischemic tissue. These results also highlight the concept that adipose lineage cells represent a suitable new cell source for therapeutic angiogenesis in ischemic disease.
Collapse
Affiliation(s)
- Valérie Planat-Benard
- Unité Mixte de Recherche 5018 Centre National de la Recherche Scientifique, Université Paul Sabatier, Centre Hospitalier Universitaire Rangueil, Toulouse, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1434
|
Simons M, Ware JA. Therapeutic angiogenesis in cardiovascular disease. Nat Rev Drug Discov 2004; 2:863-71. [PMID: 14668807 DOI: 10.1038/nrd1226] [Citation(s) in RCA: 236] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Michael Simons
- Angiogenesis Research Center and Section of Cardiology, Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| | | |
Collapse
|
1435
|
Scheubel RJ, Zorn H, Silber RE, Kuss O, Morawietz H, Holtz J, Simm A. Age-dependent depression in circulating endothelial progenitor cells in patients undergoing coronary artery bypass grafting. J Am Coll Cardiol 2004; 42:2073-80. [PMID: 14680729 DOI: 10.1016/j.jacc.2003.07.025] [Citation(s) in RCA: 267] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The effect of patient age on circulating endothelial progenitor cells (EPCs) and their mobilization during coronary artery bypass grafting (CABG) was assessed. BACKGROUND The EPCs are able to contribute to reparative neovascularization after tissue ischemia. In experimental models, reparative neovascularization is impaired in senescent animals, but the role of EPCs in this impairment, especially in humans, is unknown. METHODS In 50 consecutive patients (43 to 80 years old) with stable coronary artery disease undergoing CABG, the numbers of EPCs and the plasma levels of interleukin (IL)-6, IL-8, IL-10, and IL-18, as well as vascular endothelial growth factor (VEGF) and placental growth factor, were determined preoperatively, after coming off bypass, and 6, 12, 24, and 72 h postoperatively. RESULTS Preoperative values of EPCs were lowered with increasing age, similar to the lowering of plasma VEGF levels. These age-associated decreases could not be explained by differences in atherosclerotic risk factors or cardiac function. Bypass surgery induced a rapid mobilization in EPCs, IL-6, IL-8, IL-10, and VEGF, with a peak 6 h postoperatively. Persistently lower levels of EPCs and VEGF throughout the observation period were observed in patients >69 years old, which could not be explained by differences in the operative procedure or inflammatory IL activation. CONCLUSIONS Despite a significant increase in EPCs and release of cytochemokines during CABG, age is a major limiting factor for mobilization of EPCs. Further studies are necessary to improve the strategies for mobilization, ex vivo expansion, and re-transplantation of EPCs in aging patients.
Collapse
Affiliation(s)
- Robert J Scheubel
- Department of Cardiothoracic Surgery, and Institute of Epidemiology, Biostatistics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | | | | | | | | | | | | |
Collapse
|
1436
|
|
1437
|
Prósper Cardoso F, Herreros González J, Alegría Ezquerra E. [Stem cells to regenerate cardiac tissue in heart failure]. Rev Esp Cardiol 2004; 56:935-9. [PMID: 14563284 DOI: 10.1016/s0300-8932(03)76987-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial regeneration is one of the most promising therapeutic strategies for heart failure patients. Many experimental studies have demonstrated that different types of stem cell can differentiate into myocardial cells and tissues necessary for regeneration of the damaged myocardium, while studies in experimental animals suggest that muscle (myoblast), bone marrow (mesenchymal, endothelial or hematopoietic progenitors) and even heart cells can help to improve heart contractility in vivo. These findings have led several groups to undertake studies in patients with myocardial infarction. However, the use of cellular therapy in clinical trials is not without controversy, mainly related with the need for better knowledge before these therapeutic strategies are used in clinical practice. Although significant enhancement of our knowledge of the processes involved is fundamental, we do not consider it unreasonable to initiate clinical trials in which specific questions are posed, whose answers will allow us to make further progress.
Collapse
|
1438
|
Kinnaird T, Stabile E, Epstein SE, Fuchs S. Current perspectives in therapeutic myocardial angiogenesis. J Interv Cardiol 2004; 16:289-97. [PMID: 14562668 DOI: 10.1034/j.1600-6143.2003.08061.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The complex mechanisms mediating the development of new blood vessels are now beginning to be unraveled. In conjunction with major biotechnology advances, this has facilitated the initiation of translational research related to a novel treatment strategy for patients with myocardial or leg ischemia due to obstructive arterial disease--therapeutic angiogenesis. At present, at least 17 clinical trials of myocardial angiogenesis have been presented involving over 900 patients. Uncertainty exists as to the optimal delivery route and angiogenic agent, and this uncertainty is reflected in the diverse methodology of the trials published thus far. The majority of patients received an angiogenic protein via the intracoronary route. Other delivery techniques--such as direct intramyocardial injection via transepicardial or transendocardial routes--and other angiogenic agents, including master genes, have also been studied. Most recently, interest has grown in the potential angiogenesis effects of cell therapy--such as autologous bone marrow cells or cultured stem cells--and there are now several groups initiating Phase I/II trials in this area. This review summarizes the current evidence pertaining to the safety, feasibility, and efficacy of various angiogenic techniques aimed at enhancing myocardial blood flow and alleviating angina.
Collapse
Affiliation(s)
- Tim Kinnaird
- Cardiovascular Research Institute, MedStar Research Institute, Washington Hospital Center, 110 Irving St. NW, Suite 4B-1, Washington, DC 20010, USA.
| | | | | | | |
Collapse
|
1439
|
Silva GV, Perin EC, Dohmann HFR, Borojevic R, Silva SA, Sousa ALS, Assad JAR, Vaughn WK, Mesquita CT, Belém L, Carvalho AC, Dohmann HJF, Barroso do Amaral E, Coutinho J, Branco R, Oliveira E, Willerson JT. Catheter-based transendocardial delivery of autologous bone-marrow-derived mononuclear cells in patients listed for heart transplantation. Tex Heart Inst J 2004; 31:214-219. [PMID: 15562839 PMCID: PMC521759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Growing evidence suggests that transplantation of autologous bone-marrow mononuclear cells (ABMMNCs) can improve the perfusion and contractile function of ischemic myocardium. This procedure could potentially benefit transplant candidates awaiting a donor heart. To study the safety and feasibility of ABMMNC injection, we performed a prospective, nonrandomized, open-label study in 5 heart transplant candidates with severe ischemic heart failure. Each patient underwent baseline single-photon emission computed tomography, a ramp treadmill protocol, 2-dimensional echocardiography, 24-hour Holter monitoring, and signal-averaged electrocardiography, which were repeated at 2 and 6 months. Transendocardial delivery of ABMMNCs was done with the aid of electromechanical mapping to identify viable myocardium. Each patient received 15 ABMMNC injections of 0.2 cc each. There were no deaths, significant arrhythmias, or other major complications. The ABMMNC injection reduced the amount of ischemic myocardium (not statistically significant). More important, exercise test results improved significantly. Myocardial volume oxygen consumption increased from 10.6 +/- 3 mL/kg/min (baseline) to 16.3 +/- 7 mL/kg/min (2 months) and 23 +/- 7 mL/kg/min (6 months) (P = 0.0091). In 4 of the 5 cases, this was such an improvement that the patients were no longer eligible for cardiac transplantation. In addition, metabolic equivalents improved from 3.03 +/- 0.66 (baseline) to 4.65 +/- 1.99 (2 months) and 6.5 +/- 2.0 (6 months) (P = 0.0092). In conclusion, ABMMNC injections were performed safely and resulted in improved exercise capacity. This technique may hold promise as an alternative to medical management in patients with severe ischemic heart failure who are ineligible for conventional revascularization.
Collapse
Affiliation(s)
- Guilherme V Silva
- The Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1440
|
Rissanen TT, Rutanen J, Ylä-Herttuala S. Gene Transfer for Therapeutic Vascular Growth in Myocardial and Peripheral Ischemia. ADVANCES IN GENETICS 2004; 52:117-64. [PMID: 15522734 DOI: 10.1016/s0065-2660(04)52004-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic vascular growth in the treatment of peripheral and myocardial ischemia has not yet fulfilled its expectations in clinical trials. Randomized, double-blinded placebo-controlled trials have predominantly shown the safety and feasibility but not the clear-cut clinically relevant efficacy of angiogenic gene or recombinant growth factor therapy. It is likely that growth factor levels achieved with single injections of recombinant protein or naked plasmid DNA are too low to induce any relevant angiogenic effects. Also, the route of administration of gene transfer vectors has not been optimal in many cases leading to low gene-transfer efficacy. Animal experiments using intramuscular or intramyocardial injections of adenovirus encoding vascular endothelial growth factor (VEGF, VEGF-A), the mature form of VEGF-D, and fibroblast growth factors (FGF-1, -2, and -4) have shown high angiogenic efficacy. Adenoviral overexpression of VEGF receptor-2 ligands, VEGF-A and the mature form of VEGF-D, enlarge the preexisting capillaries in skeletal muscle and myocardium via nitric oxide(NO)-mediated mechanisms and via proliferation of both endothelial cells and pericytes, resulting in markedly increased tissue perfusion. VEGF also enhances collateral growth, which is probably secondary to increased peripheral capillary blood flow and shear stress. As a side effect of VEGF overexpression and rapid microvessel enlargement, vascular permeability increases and may result in substantial tissue edema and pericardial effusion in the heart. Because of the transient adenoviral gene expression, the majority of angiogenic effects and side effects return to baseline by 2 weeks after the gene transfer. In contrast, VEGF overexpression lasting over 4 weeks has been shown to induce the growth of a persistent vascular network in preclinical models. To improve efficacy, the choice of the vascular growth factor, gene transfer vector, and route of administration should be optimized in future clinical trials. This review is focused on these issues.
Collapse
Affiliation(s)
- Tuomas T Rissanen
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute, Kuopio University, Kuopio, Finland
| | | | | |
Collapse
|
1441
|
Mathews V, Hanson PT, Ford E, Fujita J, Polonsky KS, Graubert TA. Recruitment of bone marrow-derived endothelial cells to sites of pancreatic beta-cell injury. Diabetes 2004; 53:91-8. [PMID: 14693702 DOI: 10.2337/diabetes.53.1.91] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial progenitor cells (EPCs) are detectable in the blood and bone marrow throughout life. These cells contribute to new blood vessel formation (neovascularization) in physiological states such as wound healing and in pathological states such as tumor angiogenesis. We hypothesized that bone marrow-derived EPCs could play a role in the response to pancreatic islet cell injury. We used a murine model of experimentally induced beta-cell injury followed by transplantation with genetically marked bone marrow cells. Bone marrow-derived cells were detectable throughout the pancreas after transplantation. Whereas the total number of bone marrow-derived cells in the pancreas decreased over time, the frequency of endothelial cells (of both donor and recipient origin) increased after transplantation in the animals in which beta-cell injury had been induced. There was no evidence in this model that bone marrow-derived cells differentiated into insulin-expressing cells. This study provides evidence that bone marrow-derived EPCs are recruited to the pancreas in response to islet injury. EPC-mediated neovascularization of the pancreas could in principle be exploited to facilitate the recovery of non-terminally injured beta-cells or to improve the survival and/or function of islet allografts.
Collapse
Affiliation(s)
- Vikram Mathews
- Department of Internal Medicine, Division of Oncology, Stem Cell Biology Section, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
1442
|
Loomans CJM, de Koning EJP, Staal FJT, Rookmaaker MB, Verseyden C, de Boer HC, Verhaar MC, Braam B, Rabelink TJ, van Zonneveld AJ. Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes 2004; 53:195-9. [PMID: 14693715 DOI: 10.2337/diabetes.53.1.195] [Citation(s) in RCA: 658] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes is associated with reduced vascular repair, as indicated by impaired wound healing and reduced collateral formation in ischemia. Recently, endothelial progenitor cells (EPCs) have been identified as important regulators of these processes. We therefore explored the concept that EPCs are dysfunctional in diabetes. The number of EPCs obtained from type 1 diabetic patients in culture was 44% lower compared with age- and sex-matched control subjects (P < 0.001). This reduction was inversely related to levels of HbA(1c) (R = -0.68, P = 0.01). In addition, we demonstrated that patient EPCs were also impaired in function using an in vitro angiogenesis assay. Conditioned media from patient EPCs were significantly reduced in their capacity to support endothelial tube formation in comparison to control EPCs. Therefore, despite culturing the EPCs under normoglycemic conditions, functional differences between patient and control EPCs were maintained. Our findings demonstrate that adverse metabolic stress factors in type 1 diabetes are associated with reduced EPC numbers and angiogenicity. We hypothesize that EPC dysfunction contributes to the pathogenesis of vascular complications in type 1 diabetes.
Collapse
Affiliation(s)
- Cindy J M Loomans
- Department of Vascular Medicine and Diabetology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1443
|
Perspectivas futuras de tratamiento en la insuficiencia cardíaca: del trasplante de células a la regeneración cardíaca. Rev Esp Cardiol (Engl Ed) 2004. [DOI: 10.1016/s0300-8932(04)77226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
1444
|
Lim DS. Stem Cell Therapy for Ischemic Heart Disease : A Status Report. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2004. [DOI: 10.5124/jkma.2004.47.10.926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Do Sun Lim
- Department of Internal Medicine, Korea University College of Medicine, Anam Hospital, Korea.
| |
Collapse
|
1445
|
Regeneración miocárdica mediante la implantación intracoronaria de células madre en el infarto agudo de miocardio. Rev Esp Cardiol 2004. [DOI: 10.1016/s0300-8932(04)77091-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
1446
|
Abstract
Long before their existence was proven, work with blood islands pointed to the existence of hemangioblasts in the embryo, and it was widely accepted that such cells existed. In contrast, though evidence for adult hemangioblasts appeared at least as early as 1932, until quite recently, it was commonly assumed that there were no adult hemangioblasts. Over the past decade, these views have changed, and it is now generally accepted that a subset of bone marrow cells or their progeny can and do function as adult hemangioblasts. This chapter will examine the basic biology of bone marrow-derived hemangioblasts and endothelial cell progenitors (angioblasts) and the relationship of these adult cells to their embryonic counterparts. Efforts to define the endothelial cell progenitor phenotype will also be discussed, though to date, there is no consensus on the definitive adult phenotype, probably because there are multiple phenotypes and because the cells are plastic. Also examined are the putative roles of bone marrow-derived cells in vascular homeostasis and repair, including both their ability to differentiate and contribute directly to vascular repair, as well as to promote vascular growth by secreting pro-angiogenic factors. Finally, the use of bone marrow cells as therapeutic tools will be addressed.
Collapse
Affiliation(s)
- Gina C Schatteman
- Department of Exercise Science, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
1447
|
Candell Riera J. El implante de células madre es factible en el ámbito clínico, pero ¿será eficaz? Rev Esp Cardiol (Engl Ed) 2004. [DOI: 10.1016/s0300-8932(04)77088-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
1448
|
Obradović S, Rusović S, Balint B, Ristić-Andelkov A, Romanović R, Baskot B, Vojvodić D, Gligić B. Autologous bone marrow-derived progenitor cell transplantation for myocardial regeneration after acute infarction. VOJNOSANIT PREGL 2004; 61:519-29. [PMID: 15551805 DOI: 10.2298/vsp0405519o] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background. Experimental and first clinical studies suggest that the transplantation of bone marrow derived, or circulating blood progenitor cells, may beneficially affect postinfarction remodelling processes after acute myocardial infarction. Aim. This pilot trial reports investigation of safety and feasibility of autologous bone marrow-derived progenitor cell therapy for faster regeneration of the myocardium after infarction. Methods and results. Four male patients (age range 47-68 years) with the first extensive anterior, ST elevation, acute myocardial infarction (AMI), were treated by primary angioplasty. Bone marrow mononuclear cells were administered by intracoronary infusion 3-5 days after the infarction. Bone marrow was harvested by multiple aspirations from posterior cristae iliacae under general anesthesia, and under aseptic conditions. After that, cells were filtered through stainless steel mesh, centrifuged and resuspended in serum-free culture medium, and 3 hours later infused through the catheter into the infarct-related artery in 8 equal boluses of 20 ml. Myocardial viability in the infarcted area was confirmed by dobutamin stress echocardiography testing and single-photon emission computed tomography (SPECT) 10-14 days after infarction. One patient had early stent thrombosis immediately before cell transplantation, and was treated successfully with second angioplasty. Single average ECG revealed one positive finding at discharge, and 24-hour Holter ECG showed only isolated ventricular ectopic beats during the follow-up period. Early findings in two patients showed significant improvement of left ventricular systolic function 3 months after the infarction. There were no major cardiac events after the transplantation during further follow-up period (30-120 days after infarction). Control SPECT for the detection of ischemia showed significant improvement in myocardial perfusion in two patients 4 months after the infarction. Echocardiographic assessment in these two patients also showed significant improvement of systolic function three months after the infarction. Conclusion. Preliminary results of the study showed that the transplantation of bone marrow-derived progenitor cells into the infarcted area was safe, and feasible, and might improve myocardial function. Further follow-up will show if this treatment is effective in preventing negative remodeling of the left ventricle and reveal potential late adverse events (arrhythmogenicity and propensity for restenosis).
Collapse
Affiliation(s)
- Slobodan Obradović
- Military Medical Academy, Clinic of Emergency Medicine, Belgrade, Serbia & Montenegro.
| | | | | | | | | | | | | | | |
Collapse
|
1449
|
Affiliation(s)
- Masaaki Ii
- Division of Cardiovascular Research, Caritas St Elizabeth's Medical Center, University, School of Medicine, Boston, Mass 02135, USA
| | | |
Collapse
|
1450
|
Laufs U, Werner N, Link A, Endres M, Wassmann S, Jürgens K, Miche E, Böhm M, Nickenig G. Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation 2003; 109:220-6. [PMID: 14691039 DOI: 10.1161/01.cir.0000109141.48980.37] [Citation(s) in RCA: 567] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The molecular mechanisms by which physical training improves peripheral and coronary artery disease are poorly understood. Bone marrow-derived endothelial progenitor cells (EPCs) are thought to exert beneficial effects on atherosclerosis, angiogenesis, and vascular repair. METHODS AND RESULTS To study the effect of physical activity on the bone marrow, EPCs were quantified by fluorescence-activated cell sorter analysis in mice randomized to running wheels (5.1+/-0.8 km/d, n=12 to 16 per group) or no running wheel. Numbers of EPCs circulating in the peripheral blood of trained mice were enhanced to 267+/-19%, 289+/-22%, and 280+/-25% of control levels after 7, 14, and 28 days, respectively, accompanied by a similar increase of EPCs in the bone marrow and EPCs expanded from spleen-derived mononuclear cells. eNOS-/- mice and wild-type mice treated with N(G)-nitro-l-arginine methyl ester showed lower EPC numbers at baseline and a significantly attenuated increase of EPC in response to physical activity. Exercise NO dependently increased serum levels of vascular endothelial growth factor and reduced the rate of apoptosis in spleen-derived EPCs. Running inhibited neointima formation after carotid artery injury by 22+/-2%. Neoangiogenesis, as assessed in a subcutaneous disc model, was increased by 41+/-16% compared with controls. In patients with stable coronary artery disease (n=19), moderate exercise training for 28 days led to a significant increase in circulating EPCs and reduced EPC apoptosis. CONCLUSIONS Physical activity increases the production and circulating numbers of EPCs via a partially NO-dependent, antiapoptotic effect that could potentially underlie exercise-related beneficial effects on cardiovascular diseases.
Collapse
Affiliation(s)
- Ulrich Laufs
- Medizinische Klinik und Poliklinik der Universität des Saarlandes, Innere Medizin III, Homburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|