51
|
van Oostrom MC, van Oostrom O, Quax PHA, Verhaar MC, Hoefer IE. Insights into mechanisms behind arteriogenesis: what does the future hold? J Leukoc Biol 2008; 84:1379-91. [DOI: 10.1189/jlb.0508281] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
52
|
Chappell JC, Song J, Klibanov AL, Price RJ. Ultrasonic microbubble destruction stimulates therapeutic arteriogenesis via the CD18-dependent recruitment of bone marrow-derived cells. Arterioscler Thromb Vasc Biol 2008; 28:1117-22. [PMID: 18403725 DOI: 10.1161/atvbaha.108.165589] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We have previously shown that, under certain conditions, ultrasonic microbubble destruction creates arteriogenesis and angiogenesis in skeletal muscle. Here, we tested whether this neovascularization response enhances hyperemia in a rat model of arterial insufficiency and is dependent on the recruitment of bone marrow-derived cells (BMDCs) to treated tissues via a beta2 integrin (CD18)-dependent mechanism. METHODS AND RESULTS Sprague-Dawley rats, C57BL/6 wild-type mice, and C57BL/6 chimeric mice engrafted with BMDCs from either GFP+ or CD18-/- mice received bilateral femoral artery ligations. Microbubbles (MBs) were intravenously injected, and one gracilis muscle was exposed to pulsed 1 MHz ultrasound (US). Rat hindlimbs exhibited significant increases in adenosine-induced hyperemia and arteriogenesis compared to contralateral controls at 14 and 28 days posttreatment. US-MB-treated wild-type C57BL/6 mice exhibited significant arteriogenesis, angiogenesis, and CD11b+ monocyte recruitment; however, these responses were all completely blocked in CD18-/- chimeric mice. The number of BMDCs increased in US-MB-treated muscles of GFP+ chimeric mice; however, GFP+ BMDCs did not incorporate into microvessels as vascular cells. CONCLUSIONS In skeletal muscle affected by arterial occlusion, arteriogenesis and hyperemia can be significantly enhanced by ultrasonic MB destruction. This response depends on the recruitment, but not vascular incorporation, of BMDCs via a CD18-dependent mechanism.
Collapse
Affiliation(s)
- John C Chappell
- Department of Biomedical Engineering, University of Virginia, UVA Health System, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
53
|
van Weel V, van Tongeren RB, van Hinsbergh VWM, van Bockel JH, Quax PHA. Vascular growth in ischemic limbs: a review of mechanisms and possible therapeutic stimulation. Ann Vasc Surg 2008; 22:582-97. [PMID: 18504100 DOI: 10.1016/j.avsg.2008.02.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 01/15/2008] [Accepted: 02/29/2008] [Indexed: 01/13/2023]
Abstract
Stimulation of vascular growth to treat limb ischemia is promising, and early results obtained from uncontrolled clinical trials using angiogenic agents, e.g., vascular endothelial growth factor, led to high expectations. However, negative results from recent placebo-controlled trials warrant further research. Here, current insights into mechanisms of vascular growth in the adult, in particular the role of angiogenic factors, the immune system, and bone marrow, were reviewed, together with modes of its therapeutic stimulation and results from recent clinical trials. Three concepts of vascular growth have been described to date-angiogenesis, vasculogenesis, and arteriogenesis (collateral artery growth)-which represent different aspects of an integrated process. Stimulation of arteriogenesis seems clinically most relevant and has most recently been attempted using autologous bone marrow transplantation with some beneficial results, although the mechanism of action is not completely understood. Better understanding of the highly complex molecular and cellular mechanisms of vascular growth may yet lead to meaningful clinical applications.
Collapse
Affiliation(s)
- V van Weel
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | |
Collapse
|
54
|
Schirmer SH, Fledderus JO, Bot PT, Moerland PD, Hoefer IE, Baan J, Henriques JP, van der Schaaf RJ, Vis MM, Horrevoets AJ, Piek JJ, van Royen N. Interferon-β Signaling Is Enhanced in Patients With Insufficient Coronary Collateral Artery Development and Inhibits Arteriogenesis in Mice. Circ Res 2008; 102:1286-94. [DOI: 10.1161/circresaha.108.171827] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Stimulation of collateral artery growth in patients has been hitherto unsuccessful, despite promising experimental approaches. Circulating monocytes are involved in the growth of collateral arteries, a process also referred to as arteriogenesis. Patients show a large heterogeneity in their natural arteriogenic response on arterial obstruction. We hypothesized that circulating cell transcriptomes would provide mechanistic insights and new therapeutic strategies to stimulate arteriogenesis. Collateral flow index was measured in 45 patients with single-vessel coronary artery disease, separating collateral responders (collateral flow index, >0.21) and nonresponders (collateral flow index, ≤0.21). Isolated monocytes were stimulated with lipopolysaccharide or taken into macrophage culture for 20 hours to mimic their phenotype during arteriogenesis. Genome-wide mRNA expression analysis revealed 244 differentially expressed genes (adjusted
P
, <0.05) in stimulated monocytes. Interferon (IFN)-β and several IFN-related genes showed increased mRNA levels in 3 of 4 cellular phenotypes from nonresponders. Macrophage gene expression correlated with stimulated monocytes, whereas resting monocytes and progenitor cells did not display differential gene regulation. In vitro, IFN-β dose-dependently inhibited smooth muscle cell proliferation. In a murine hindlimb model, perfusion measured 7 days after femoral artery ligation showed attenuated arteriogenesis in IFN-β–treated mice compared with controls (treatment versus control: 31.5±1.2% versus 41.9±1.9% perfusion restoration,
P
<0.01). In conclusion, patients with differing arteriogenic response as measured with collateral flow index display differential transcriptomes of stimulated monocytes. Nonresponders show increased expression of IFN-β and its downstream targets, and IFN-β attenuates proliferation of smooth muscle cells in vitro and hampers arteriogenesis in mice. Inhibition of IFN-β signaling may serve as a novel approach for the stimulation of collateral artery growth.
Collapse
Affiliation(s)
- Stephan H. Schirmer
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Joost O. Fledderus
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Pieter T.G. Bot
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Perry D. Moerland
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Imo E. Hoefer
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Jan Baan
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - José P.S. Henriques
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - René J. van der Schaaf
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Marije M. Vis
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Anton J.G. Horrevoets
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Jan J. Piek
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| | - Niels van Royen
- From the Departments of Cardiology (S.H.S., P.T.G.B., J.B., J.P.S.H., R.J.v.d.S., M.M.V., J.J.P., N.v.R.), Medical Biochemistry (J.O.F., A.J.G.H.), and Clinical Epidemiology, Biostatistics and Bioinformatics (P.D.M.), Academic Medical Center, University of Amsterdam; and Department of Experimental Cardiology (I.E.H.), University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
55
|
Paulsson JM, Dadfar E, Held C, Jacobson SH, Lundahl J. In vivo transmigrated monocytes from patients with stable coronary artery disease have a reduced expression of CD11b. Clin Exp Immunol 2008; 153:196-204. [PMID: 18460014 DOI: 10.1111/j.1365-2249.2008.03676.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Coronary artery disease (CAD) is characterized by infiltration of monocyte derived cells in the intima of the vessel wall. We hypothesized that accumulation of these cells is caused partly by an altered monocyte transmigration process in CAD. To gain insight into this issue we applied the skin blister method that allows collection of in vivo transmigrated cells at sites of local inflammation. Nineteen patients with stable CAD and 19 matched controls were enrolled. Markers of inflammation and gradients of chemokines, as well as adhesion molecule expression and up-regulation capacity, were studied. The expression of inflammatory markers, such as C-reactive protein, interleukin (IL)-6, tumour necrosis factor-alpha and IL-10, was similar in patients and controls, indicating that patients were in a stable phase of the disease. Expression of adhesion molecules, CD11b and very late activation antigen-4, on peripheral monocytes did not differ between patients and controls. However, following in vivo transmigration, monocytes in patients with CAD had a significantly reduced expression and mobilization of CD11b. The effect on CD11b could not be reproduced by in vitro stimulation with blister fluid, representing a local inflammatory milieu, or in an in vitro system of transmigration. These findings point towards differences in monocyte CD11b expression and availability at an inflammatory site between patients with CAD and healthy controls.
Collapse
Affiliation(s)
- J M Paulsson
- Department of Clinical Immunology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
56
|
Schirmer SH, van Royen N. Stimulation of collateral artery growth: a potential treatment for peripheral artery disease. Expert Rev Cardiovasc Ther 2007; 2:581-8. [PMID: 15225117 DOI: 10.1586/14779072.2.4.581] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the course of peripheral artery occlusive disease, blood flow to peripheral tissue progressively decreases in a substantial portion of patients, leading to insufficient oxygenation and to the occurrence of claudication or critical limb ischemia. Arteriogenesis (collateral artery growth) is a powerful natural mechanism by which large conductance vessels develop that circumvent sites of obstruction. Promising experimental data on both hypoxia-driven angiogenesis as well as monocyte-orchestrated arteriogenesis have raised high hopes for clinical application. Both endothelial growth factors to stimulate angiogenesis (i.e., capillary growth) and monocyte-attracting or -activating substances to stimulate arteriogenesis, have been proposed as potential new therapeutic agents. However, transferring the promising experimental results into clinical practice has been more cumbersome than initially anticipated. Some recent clinical studies are now focusing more specifically on the stimulation of arteriogenesis. This review will critically evaluate the results of preclinical and clinical investigations on the stimulation of vascular growth, focusing specifically on the peripheral circulation.
Collapse
Affiliation(s)
- Stephan H Schirmer
- Department of Internal Medicine III-Cardiology and Angiology, University Hospital Freiburg, Germany.
| | | |
Collapse
|
57
|
Abstract
Pharmacological attempts to stimulate the growth of collateral arteries (arteriogenesis) are evolving towards a new treatment option for patients with vascular occlusive diseases. This enlargement of small pre-existing anastomoses towards large conductance arteries takes place independent of local oxygen tension and is driven by changes in luminal shear stress and infiltration of circulating cells. With the increasing knowledge regarding the distinct differences between capillary sprouting (angiogenesis) and arteriogenesis, several cytokines and growth factors have been demonstrated to stimulate the growth of arterial blood vessels in preclinical models of vascular disease. However, the translation towards clinical practice remains difficult and first in-man trials show limited success. Intensive research especially regarding new drug delivery platforms and the potentially serious side effects of pro-arteriogenic therapeutics is warranted before stimulation of arteriogenesis could become a significant treatment option for vascular occlusive diseases. This review focuses on the recent advances in the field of collateral artery growth. In addition, possible means to overcome the hurdles that have hampered the clinical implementation of pro-arteriogenic therapies will be discussed.
Collapse
|
58
|
Ozaki CK, Jiang Z, Berceli SA. TNF-alpha and shear stress-induced large artery adaptations. J Surg Res 2007; 141:299-305. [PMID: 17574273 PMCID: PMC2032015 DOI: 10.1016/j.jss.2006.12.563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 12/18/2006] [Accepted: 12/29/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNF-alpha) up-regulation has been associated with both low and high shear-induced arterial remodeling. To address this apparent paradox and to define the biology of TNF-alpha signaling in large arteries, we tested the hypotheses that differential temporal expression of TNF-alpha drives shear-regulated arterial remodeling. MATERIALS AND METHODS Both low- and high-shear environments in the same rabbit were surgically created for common carotid arteries. Common carotid arteries (n = 60 total) were harvested after d0, d1, d3, d7, and d14 and analyses included morphology, TNF-alpha, and IL-10 mRNA quantitation. In separate experiments, animals received pegylated soluble TNF-alpha Type 1 receptor (PEG sTNF-RI) or vehicle via either short- or long-term dosing to define the effect of TNF-alpha blockade. RESULTS The model yielded a 14-fold shear differential (P < 0.001) with medial thickening under low shear (P = 0.025), and evidence of outward remodeling with high shear (P = 0.007). Low shear immediately up-regulated TNF-alpha expression approximately 50 fold (P < 0.001) at d1. Conversely, high shear-induced delayed and sustained TNF-alpha expression (22-fold at d7, P = 0.012; 23-fold at d14, P = 0.007). Both low and high shear gradually induced IL-10 expression (P = 0.002 and P = 0.004, respectively). Neither short-term (5-day) nor long-term (14-day) blockage of TNF-alpha signaling resulted in treatment-induced changes in the remodeling of low- or high-shear arteries. CONCLUSIONS Shear stress differentially and temporally regulates TNF-alpha expression in remodeling large arteries. However, TNF-alpha blockage did not substantially impact the final shear-induced morphology, suggesting that large arteries can remodel in response to flow perturbations independent of TNF-alpha signaling.
Collapse
Affiliation(s)
- C Keith Ozaki
- University of Florida College of Medicine and the Malcom Randall VAMC, Gainesville, Florida, USA.
| | | | | |
Collapse
|
59
|
Dadfar E, Jacobson SH, Lundahl J. Newly recruited human monocytes have a preserved responsiveness towards bacterial peptides in terms of CD11b up-regulation and intracellular hydrogen peroxide production. Clin Exp Immunol 2007; 148:573-82. [PMID: 17386075 PMCID: PMC1941923 DOI: 10.1111/j.1365-2249.2007.03373.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2007] [Indexed: 01/04/2023] Open
Abstract
The transmigration of peripheral human monocytes to the interstitium is a fundamental step in the host-defence mechanism against infections. Little is known about the state of function of in vivo transmigrated interstitial monocytes prior to differentiation into macrophages and dendritic cells. We hypothesized that newly recruited interstitial monocytes have a preserved responsiveness against bacterial-related peptides, giving them a specific role in the immediate defence against invading pathogens. In order to test this hypothesis, we explored the responsiveness of in vivo transmigrated as well as peripheral monocytes, in terms of CD11b expression and H(2)O(2) production towards the bacterial-related peptide formylmethionylleucylphenylalanine (fMLP) by the use of a skin chamber technique. In addition, we analysed the concentration of interleukin (IL)-8, monocyte chemotactic protein-1 (MCP-1) and tumour necrosis factor (TNF)-alpha in the skin blister exudates and in the circulation. We demonstrate that in vivo-transmigrated monocytes had a fivefold higher CD11b expression compared to monocytes obtained from the peripheral circulation. fMLP exposure induced a significantly higher CD11b expression on transmigrated cells compared to peripheral monocytes. In addition, newly recruited monocytes had a preserved H(2)O(2) production. The interstitial concentration of IL-8, MCP-1 and TNF-alpha was significantly higher in blister exudates compared to that in the peripheral circulation. Thus, in vivo transmigrated human monocytes preserve their capacity to respond towards bacterial peptides in terms of CD11b up-regulation and H(2)O(2) generation. These data strengthen a role for newly recruited interstitial human monocytes in the immediate defence against invading pathogens.
Collapse
Affiliation(s)
- E Dadfar
- Department of Medicine, Clinical Immunology and Allergy Unit, Karolinska Institutet, Karolinska University Hospital, Sweden.
| | | | | |
Collapse
|
60
|
Voskuil M, Hoefer IE, van Royen N, Hua J, de Graaf S, Bode C, Buschmann IR, Piek JJ. Abnormal monocyte recruitment and collateral artery formation in monocyte chemoattractant protein-1 deficient mice. Vasc Med 2006; 9:287-92. [PMID: 15678621 DOI: 10.1191/1358863x04vm571oa] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Monocyte chemoattractant protein 1 (MCP-1) has been shown to be effective for the stimulation of collateral artery formation in small and large animal models. The availability of a genetic knockout mouse enables evaluation of the importance of the role of MCP-1 in the natural course of collateral artery growth. In a total of 21 MCP-1 -/- as well as 13 of the appropriate genetic background controls ([129Sv/J X C57BI/6J]F1), a femoral artery ligation was performed. Subsequently, a polyethylene catheter, connected to an osmotic minipump, was inserted retrogradely into the occluded femoral artery with the tip pointing upstream. Using this technique, PBS (MCP-1 -/-: n = 13 and C57BI/6J: n = 13) or MCP-1 (JE; MCP-1 -/-: n = 8) was delivered intra-arterially. Seven days after ligation, determination of hind limb flow was assessed by controlled tissue perfusion using differently labeled fluorescent microspheres. MCP-1 -/- mice exhibited a reduction of hind limb flow of 32.9 +/- 9.2% of the unligated hind limb, compared with 55.4 +/- 6.8% in C57BI/6J mice (p<0.01). MCP-1 -/- mice that underwent a subsequent 'rescue' treatment with MCP-1 showed a restoration of flow to a level of 47.4 +/- 9.8% (p = NS compared with PBS-treated C57BI/6J). Specific immunohistochemical staining for monocytes (MOMA-2: MCP-1 -/-, n = 5 and C57BI/6J, n = 5) showed a reduced number of monocytes around developing collateral arteries in the MCP-1 -/- mice. In conclusion, our data show that the absence of MCP-1 causes a strong reduction in flow restoration after femoral artery occlusion, coinciding with a reduced monocyte attraction, emphasizing the central role of this chemokine in the multifactorial process of collateral artery formation.
Collapse
Affiliation(s)
- Michiel Voskuil
- Department of Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Schepers A, Pires NMM, Eefting D, de Vries MR, van Bockel JH, Quax PHA. Short-term dexamethasone treatment inhibits vein graft thickening in hypercholesterolemic ApoE3Leiden transgenic mice. J Vasc Surg 2006; 43:809-15. [PMID: 16616241 DOI: 10.1016/j.jvs.2005.11.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 11/12/2005] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The aim of this study was to assess whether the anti-inflammatory agent dexamethasone can inhibit vein graft thickening without the occurrence of serious side effects. METHODS Venous interposition grafting was performed in the common carotid artery of hypercholesterolemic ApoE3Leiden transgenic mice. Mice were treated with dexamethasone (0.15 mg.kg(-1).d(-1) orally), and after 28 days, vein graft thickening was quantified. RESULTS Treatment with dexamethasone resulted in a significant 43% reduction in lesion area without changes in lesion composition when compared with nontreated controls. However, dexamethasone, when administered for a prolonged period of time, is known for its potentially serious side effects. To overcome these potential side effects of prolonged dexamethasone treatment, the effect of a short-term 7-day dexamethasone treatment was studied. This short dexamethasone treatment resulted in a 49% decrease of vein graft thickening at 28 days. Furthermore, it was demonstrated that dexamethasone treatment led to reduced local expression of several proinflammatory cytokines and factors in the vein grafts 24 hours after surgery. Finally, observations in mice were verified in human saphenous organ cultures. Exposure to dexamethasone for either 7 or 28 days significantly reduced intimal hyperplasia formation on cultured saphenous vein segments. CONCLUSIONS Short-term anti-inflammatory treatment with dexamethasone leads to a significant reduction in vein graft thickening over an extended period, possibly by the reduction of early expression of proinflammatory cytokines. This 7-day treatment minimizes the risk of unwanted side effects of long-term dexamethasone treatment and may be a new approach to prevent graft failure.
Collapse
Affiliation(s)
- Abbey Schepers
- Gaubius Laboratory, TNO Quality of Life, Leiden University Medical Center, The Netherlands
| | | | | | | | | | | |
Collapse
|
62
|
Zentilin L, Tafuro S, Zacchigna S, Arsic N, Pattarini L, Sinigaglia M, Giacca M. Bone marrow mononuclear cells are recruited to the sites of VEGF-induced neovascularization but are not incorporated into the newly formed vessels. Blood 2006; 107:3546-54. [PMID: 16391016 DOI: 10.1182/blood-2005-08-3215] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is a key regulator of blood vessel formation during both vasculogenesis and angiogenesis. The prolonged expression of VEGF in the normoperfused skeletal muscles of adult rodents after gene transfer using AAV vectors induces the formation of a large set of new capillaries and small arteries. Notably, this process is accompanied by the massive infiltration by mononuclear cells. This observation raises the possibility that these cells might represent circulating progenitors that are eventually incorporated in the newly formed vessels. Here we explore this possibility by exploiting 4 different experimental models based on (a) the transplantation of male bone marrow into female recipients; (b) the transplantation of Tie2-GFP transgenic bone marrow; (c) the transplantation of bone marrow in the presence of erythropoietin (EPO), a mobilizer of endothelial progenitor cells (EPCs); and (d) the reimplantation of ex vivo-expanded EPCs. In all 4 models, VEGF acted as a powerful attractor of bone marrow-derived mononuclear cells, bearing different myeloid and endothelial markers proper of the EPCs to the sites of neovascularization. In no case, however, were the attracted cells incorporated in the newly formed vasculature. These observations indicate that new blood vessel formation induced by VEGF occurs through bona fide sprouting angiogenesis; the contribution of the infiltrating bone marrow-derived cells to this process still remains enigmatic.
Collapse
|
63
|
Waeckel L, Bignon J, Liu JM, Markovits D, Ebrahimian TG, Vilar J, Mees B, Blanc-Brude O, Barateau V, Le Ricousse-Roussanne S, Duriez M, Tobelem G, Wdzieczak-Bakala J, Lévy BI, Silvestre JS. Tetrapeptide AcSDKP Induces Postischemic Neovascularization Through Monocyte Chemoattractant Protein-1 Signaling. Arterioscler Thromb Vasc Biol 2006; 26:773-9. [PMID: 16410461 DOI: 10.1161/01.atv.0000203510.96492.14] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND We investigated the putative proangiogenic activity and molecular pathway(s) of the tetrapeptide acetyl-N-Ser-Asp-Lys-Pro (AcSDKP) in a model of surgically induced hindlimb ischemia. METHODS AND RESULTS Hindlimb ischemia was induced by femoral artery ligature and an osmotic minipump was implanted subcutaneously to deliver low (0.12 mg/kg per day) or high (1.2 mg/kg per day) doses of AcSDKP, for 7 or 21 days. Angiography scores, arteriole density, capillary number, and foot perfusion were increased at day 21 in the high-dose AcSDKP-treated mice (by 1.9-, 1.8-, 1.3-, and 1.6-fold, respectively) compared with control animals (P<0.05, P<0.01, P<0.01, respectively). AcSDKP treatment for 24 hours upregulated the monocyte chemoattractant protein-1 (MCP-1) mRNA and protein levels by 1.5-fold in cultured endothelial cells (P<0.01). In the ischemic hindlimb model, administration of AcSDKP also enhanced MCP-1 mRNA levels by 90-fold in ischemic leg (P<0.001) and MCP-1 plasma levels by 3-fold (P<0.001 versus untreated ischemic control mice). MCP-1 levels upregulation were associated with a 2.3-fold increase in the number of Mac3-positive cells in ischemic area of AcSDKP-treated mice (P<0.001 versus untreated animals). Interestingly, AcSDKP-induced monocyte/macrophage infiltration and postischemic neovascularization was fully blunted in MCP-1-deficient animals. CONCLUSIONS AcSDKP stimulates postischemic neovascularization through activation of a proinflammatory MCP-1-related pathway.
Collapse
Affiliation(s)
- Ludovic Waeckel
- Cardiovascular Research Center, INSERM U689, Université Paris 7, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Hoefer IE, van Royen N, Jost MM. Experimental models of arteriogenesis: differences and implications. Lab Anim (NY) 2006; 35:36-44. [PMID: 16446736 DOI: 10.1038/laban0206-36] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 08/22/2005] [Indexed: 01/23/2023]
Abstract
Cardiovascular and cerebrovascular disease represent the two most common causes of mortality and morbidity in western countries, and the treatment for these is generally by the mechanical restoration of blood flow in the affected tissues. Stimulation of collateral artery growth (arteriogenesis) provides a potential alternative option for the treatment of patients suffering from occlusive artery disease. Therefore, researchers have established several angiogenesis and arteriogenesis animal models to investigate basic mechanisms and pharmacological modulation of collateral artery growth. The authors highlight the most important aspects of vascular growth, discuss different methods and techniques for examining the process, and review the advantages and disadvantages associated with the animal models available for studying this phenomenon.
Collapse
Affiliation(s)
- Imo E Hoefer
- Department of Experimental Cardiology, UMC, University of Utrecht, The Netherlands.
| | | | | |
Collapse
|
65
|
Shireman PK, Contreras-Shannon V, Reyes-Reyna SM, Robinson SC, McManus LM. MCP-1 parallels inflammatory and regenerative responses in ischemic muscle. J Surg Res 2006; 134:145-57. [PMID: 16488443 DOI: 10.1016/j.jss.2005.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Revised: 11/30/2005] [Accepted: 12/05/2005] [Indexed: 11/17/2022]
Abstract
BACKGROUND Monocyte chemotactic protein-1 (MCP-1) is important in macrophage recruitment and activation. However, the magnitude and temporal sequence of MCP-1 expression in relation to tissue injury and regeneration following ischemic injury remains unknown. MATERIALS AND METHODS Hind limb ischemia was induced by femoral artery excision (FAE) in C57Bl/6J mice; a sham surgery was performed on the contralateral leg. Muscle lysates were used to measure MCP-1 and activities of creatine kinase, lactate dehydrogenase, and myeloperoxidase. Histology and immunohistochemistry were used to localize inflammation and MCP-1. RESULTS FAE resulted in a prolonged period of ischemia and the administration of MCP-1 did not alter the restoration of perfusion. One day after femoral artery excision, extensive muscle necrosis and neutrophils were prevalent throughout the musculature of the lower leg. By 3 days, a mononuclear cell infiltrate predominated in association with robust muscle regeneration as indicated by myoD expression. Concomitantly, myeloperoxidase was maximally increased. Muscle enzymes (creatine kinase and lactate dehydrogenase) were maximally decreased within 3 days and returned to baseline levels by day 14, a time course consistent with injury and regeneration observed by histology. In parallel with these inflammatory and regenerative events, MCP-1 in muscle was maximally increased at day 3. By immunohistochemistry, MCP-1 was within vascular endothelial cells and infiltrating macrophages in areas of ischemic injury. CONCLUSIONS The transient increases and selective tissue distribution of MCP-1 during early inflammation and muscle regeneration support the hypothesis that this cytokine participates in the early reparative events preceding the restoration of vascular perfusion following ischemic injury.
Collapse
Affiliation(s)
- Paula K Shireman
- South Texas Veterans Health Care System, San Antonio, TX 78229-3900, USA.
| | | | | | | | | |
Collapse
|
66
|
Witzenbichler B, Mahfoudi A, Soubrier F, Le Roux A, Branellec D, Schultheiss HP, Isner JM. Intramuscular gene transfer of fibroblast growth factor-1 using improved pCOR plasmid design stimulates collateral formation in a rabbit ischemic hindlimb model. J Mol Med (Berl) 2005; 84:491-502. [PMID: 16389546 DOI: 10.1007/s00109-005-0031-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2005] [Accepted: 11/17/2005] [Indexed: 11/29/2022]
Abstract
Fibroblast growth factor 1 (FGF1) is an angiogenic factor known to play a role in the growth of arteries. The purpose of this study was to evaluate the usefulness of direct intramuscular injection of an optimized expression plasmid encoding FGF1 to augment collateral formation and tissue perfusion in a rabbit ischemic hindlimb model. Truncated FGF1 fused to the human fibroblast interferon (FIN) signal peptide was expressed from a newly designed plasmid backbone with an improved safety profile for gene therapy applications. In vitro, optimization of plasmid design yielded in a dramatic increase in expression efficiency for FGF1, independent of the presence of a signal peptide, as analyzed by Western Blotting. In vivo, successful transgene expression could be demonstrated by FGF1 immunostaining after gene application. FGF1 plasmid containing FIN signal peptide (100, 500, and 1,000 mug), when injected into ischemic muscle areas of rabbits 10 days after ligation of the external iliac artery, exhibited a pronounced therapeutic effect on collateral formation to the ischemic hindlimb in a dose-depending manner, as assessed by physiological (blood pressure ratio, maximal intra-arterial Doppler flow) and anatomical (angiographic score, histologic evaluation of capillary density) measurements 30 days after therapy, compared to saline or lacZ control plasmid. FGF1 plasmid without a signal peptide sequence resulted in a comparable therapeutic effect on collateral formation at comparable doses (500 and 1,000 mug). Our results indicate that intramuscular FGF1 gene application could be useful to stimulate collateral formation in a situation of chronic peripheral ischemia. The presence of a signal peptide does not seem to be obligatory to achieve bioactivity of intramuscular transfected FGF1. An optimized vector design improved both biosafety of gene transfer and expression efficiency of the transgene, rendering this vector highly suitable for human gene therapy. Therefore, this new generation vector encoding FGF1 might be useful as an alternative treatment for patients with chronic ischemic disorders not amenable to conventional therapy.
Collapse
Affiliation(s)
- Bernhard Witzenbichler
- Department of Cardiology and Pneumology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
67
|
Zbinden S, Zbinden R, Meier P, Windecker S, Seiler C. Safety and efficacy of subcutaneous-only granulocyte-macrophage colony-stimulating factor for collateral growth promotion in patients with coronary artery disease. J Am Coll Cardiol 2005; 46:1636-42. [PMID: 16256861 DOI: 10.1016/j.jacc.2005.01.068] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2004] [Revised: 01/19/2005] [Accepted: 01/25/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVES This study was designed to investigate the safety and efficacy of a short-term subcutaneous-only granulocyte-macrophage colony-stimulating factor (GM-CSF) protocol for coronary collateral growth promotion. BACKGROUND The safety and efficacy of an exclusively systemic application of GM-CSF in patients with coronary artery disease (CAD) and collateral artery promotion has not been studied so far. METHODS In 14 men (age 61 +/- 11 years) with chronic stable CAD, the effect of GM-CSF (molgramostim) on quantitatively assessed collateral flow was tested in a randomized, double-blind, placebo-controlled fashion. The study protocol consisted of an invasive collateral flow index (CFI) measurement in a stenotic as well as a normal coronary artery before and after a two-week period with subcutaneous GM-CSF (10 microg/kg; n = 7) or placebo (n = 7). Collateral flow index was determined by simultaneous measurement of mean aortic, distal coronary occlusive, and central venous pressure. RESULTS Collateral flow index in all vessels changed from 0.116 +/- 0.05 to 0.159 +/- 0.07 in the GM-CSF group (p = 0.028) and from 0.166 +/- 0.06 to 0.166 +/- 0.04 in the placebo group (p = NS). The treatment-induced difference in CFI was +0.042 +/- 0.05 in the GM-CSF group and -0.001 +/- 0.04 in the placebo group (p = 0.035). Among 11 determined cytokines, chemokines, and their monocytic receptor concentrations, the treatment-induced change in CFI was predicted by the respective change in tumor necrosis factor-alpha concentration. Two of seven patients in the GM-CSF group and none in the placebo group suffered an acute coronary syndrome during the treatment period. CONCLUSIONS A subcutaneous-only, short-term protocol of GM-CSF is effective in promoting coronary collateral artery growth among patients with CAD. However, the drug's safety regarding the occurrence of acute coronary syndrome is questionable.
Collapse
Affiliation(s)
- Stephan Zbinden
- Department of Cardiology, University Hospital, Bern, Switzerland
| | | | | | | | | |
Collapse
|
68
|
Pakala R, Stabile E, Jang GJ, Clavijo L, Waksman R. Rapamycin Attenuates Atherosclerotic Plaque Progression in Apolipoprotein E Knockout Mice. J Cardiovasc Pharmacol 2005; 46:481-6. [PMID: 16160601 DOI: 10.1097/01.fjc.0000177985.14305.15] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Rapamycin has been shown to reduce neointimal thickening in the setting of balloon angioplasty and chronic graft vessel disease. This study was designed to test the effect of oral rapamycin on atherosclerotic plaque progression and the possible mechanism involved. Apolipoprotein E (apoE) knockout mice were fed either a diet supplemented with cholesterol or with cholesterol and rapamycin. At 4 and 8 weeks, quantitative analyses of plaque area and macrophage numbers were determined. Plasma cholesterol, triglyceride, and whole-blood rapamycin levels were measured. Rapamycin could be detected in the blood of mice (117+/-7 pg/mL). In mice fed with rapamycin, atherosclerotic lesions covered 22% of the aortic arch as compared with 41% in cholesterol-fed mice. The macrophage count was significantly lower in the rapamycin-fed mice as compared with cholesterol-fed mice. Rapamycin, in a dose-dependent manner, inhibited monocyte chemotaxis elicited by stromal cell-derived factor-1. Lesions in the cholesterol-fed mice had complex atherosclerotic plaque with acellular core, cholesterol clefts, and an abundant collection of monocytes/macrophages. Lesions in the rapamycin-fed mice were mainly composed of monocytes/macrophages. Oral rapamycin is effective in slowing the progression of atherosclerosis. Along with its multitude actions, attenuation of monocyte chemotaxis may be one more way by which rapamycin attenuates plaque progression.
Collapse
Affiliation(s)
- Rajbabu Pakala
- Cardiovascular Research Institute, Washington Hospital Center, Washington, DC 20010, USA
| | | | | | | | | |
Collapse
|
69
|
Chappell JC, Klibanov AL, Price RJ. Ultrasound-microbubble-induced neovascularization in mouse skeletal muscle. ULTRASOUND IN MEDICINE & BIOLOGY 2005; 31:1411-22. [PMID: 16223645 DOI: 10.1016/j.ultrasmedbio.2005.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 06/09/2005] [Accepted: 06/14/2005] [Indexed: 05/04/2023]
Abstract
Ultrasound-microbubble (US-MB) interactions stimulate neovascularization in rat gracilis muscle (GM). We examined microvascular remodeling (MVR) in GMs of C57BL/6 and balb/C mice following ultrasonic MB destruction. A range of MB dosages were administered IV, and exposed GMs received US. Muscles harvested 3, 7 and 14 d posttreatment were stained for vascular markers and assessed for changes in microvessel number, diameter and length. Muscles receiving a low MB dose (LMBD) and US showed significant increases in microvascular density after 3 d, returning to sham levels after one week. A MB dose producing maximum capillary disruptions was then established. This high MB dose (HMBD) facilitated significant MVR in C57BL/6 mice after one week. Balb/C GMs exhibited neovascularization 3 d, but not 7 or 14 d, following US-HMBD treatment. We conclude that HMBD in C57BL/6 mice induces a more sustained neovascularization response compared to balb/C or LMBD-treated C57BL/6 muscles; however, this response is still impermanent.
Collapse
Affiliation(s)
- John C Chappell
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | | | | |
Collapse
|
70
|
Lo IC, Shih JM, Jiang MJ. Reactive oxygen species and ERK 1/2 mediate monocyte chemotactic protein-1-stimulated smooth muscle cell migration. J Biomed Sci 2005; 12:377-88. [PMID: 15917991 DOI: 10.1007/s11373-005-1703-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Accepted: 12/20/2004] [Indexed: 10/25/2022] Open
Abstract
Monocyte chemotactic protein-1 (MCP-1), a potent chemoattractant for monocytes, is thought to play a major role in atherosclerosis, but whether its atherogenic effects involve the direct modulation of vascular smooth muscle cell (SMC) functions remains unclear. This study examined the effects of MCP-1 on the migration of cultured A7r5 SMCs and the signaling pathways involved. Addition of recombinant MCP-1 stimulated SMC migration in modified Boyden chambers coated with type I collagen in a concentration-dependent manner, with 10(-9) M being maximally effective. Using untreated A7r5 cells, two MCP-1 receptors, CCR2 and CCR4, were detected and MCP-1 secretion was significantly increased by stimulation with platelet-derived growth factor. MCP-1-stimulated A7r5 migration was completely blocked by the NAD(P)H oxidase inhibitor, diphenylene iodonium (DPI), and dose-dependently inhibited by polyethylene glycol-conjugated superoxide dismutase (PEG-SOD), suggesting a role for reactive oxygen species (ROS) in this process. During MCP-1 stimulation, ROS production increased rapidly, then gradually decayed over 60 min, and this effect was markedly decreased by pretreatment with DPI or PEG-SOD. Interestingly, U0126 and PD98059, which inhibit activation of extracellular signal-regulated kinases 1/2 (ERK 1/2), significantly inhibited MCP-1-activated ROS generation. Furthermore, transfection of an active mutant of MEK1 (ERK 1/2 kinase) markedly increased superoxide production in rat aortic smooth muscle cells, as detected by dihydroethydium staining, suggesting that ERK 1/2 activation stimulates ROS generation. ERK 1/2 activation was increased for at least 30 min in cells incubated with MCP-1, and this effect was abolished by U0126 or DPI pretreatment. These results demonstrate that MCP-1 is a chemoattractant for SMCs and that MCP-1-stimulated migration requires both ROS production and ERK 1/2 activation in a positive activation loop, which may contribute to the atherogenic effects of MCP-1.
Collapse
Affiliation(s)
- I-Chung Lo
- Department of Cell Biology and Anatomy, National Cheng Kung University Medical College, Taiwan
| | | | | |
Collapse
|
71
|
Hoefer IE, Grundmann S, van Royen N, Voskuil M, Schirmer SH, Ulusans S, Bode C, Buschmann IR, Piek JJ. Leukocyte subpopulations and arteriogenesis: Specific role of monocytes, lymphocytes and granulocytes. Atherosclerosis 2005; 181:285-93. [PMID: 16039282 DOI: 10.1016/j.atherosclerosis.2005.01.047] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 01/21/2005] [Accepted: 01/27/2005] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Circulating leukocytes play a crucial role during arteriogenesis. However, known pro-arteriogenic compounds (MCP-1, GM-CSF) acting via monocytic pathways also exert positive effects on granulocytes and lymphocytes. The role of these two cell types in arteriogenesis remains yet to be clarified, which was the aim of the current study. METHODS Ninety New Zealand White Rabbits received either phosphate buffered saline (PBS), monocyte chemoattractant protein-1 (MCP-1), interleukin-8 (IL-8), neutrophil activating protein-2 (NAP-2) or lymphotactin (Ltn) via osmotic minipumps after unilateral femoral artery ligation. In vitro stimulation and in vivo assessment of chemoattraction confirmed cell-specific action of the compounds in rabbits. Arteriogenesis was evaluated by angiography and collateral conductance measurements using fluorescent microspheres. Quantitative immunohistology was used to quantify transmigrated leukocyte subtypes after infusion of the factors. RESULTS MCP-1 infusion attracts monocytes and granulocytes, whereas IL-8 attracts all three cell types albeit monocytes to a significantly lower degree than MCP-1. NAP-2 and lymphotactin selectively attract granulocytes, respectively, lymphocytes. Of the tested cytokines, only MCP-1 stimulates arteriogenesis, as assessed by collateral conductance measurements ((ml/(min 100 mmHg)): PBS, 50.70+/-5.15; MCP-1, 216.30+/-12.30; IL-8, 58.91+/-5.56; NAP-2, 66.83+/-8.72; Ltn, 52.80+/-5.37) and angiographic findings. CONCLUSION This study for the first time provides evidence that not granulocytes or T-lymphocytes but monocytes are the key mediators of arteriogenesis.
Collapse
Affiliation(s)
- Imo E Hoefer
- Research Group for Experimental and Clinical Arteriogenesis, Department of Cardiology, University of Freiburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Ayabe N, Babaev VR, Tang Y, Tanizawa T, Fogo AB, Linton MF, Ichikawa I, Fazio S, Kon V. Transiently heightened angiotensin II has distinct effects on atherosclerosis and aneurysm formation in hyperlipidemic mice. Atherosclerosis 2005; 184:312-21. [PMID: 16023123 DOI: 10.1016/j.atherosclerosis.2005.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 04/22/2005] [Accepted: 05/04/2005] [Indexed: 11/22/2022]
Abstract
Experimentally sustained increase in angiotensin II (AngII) promotes tissue destruction in various cardiovascular disorders. We examined whether transiently heightened AngII affects subsequent atherosclerosis and aneurysm formation. AngII or saline was administered for 2 weeks to apolipoprotein E (apoE)-deficient mice. Mice were sacrificed at the end of the 2-week infusion or 6- or 14 weeks later. Short-term AngII did not affect atherosclerosis immediately following the infusion or 6 weeks later. By contrast, 14 weeks after infusion there was remarkably more atherosclerosis in previously AngII-exposed mice. Preceding the build up of atherosclerotic lesions, AngII-exposure increased mRNA expression and immunostaining of monocyte chemoattractant protein-1 (MCP-1) and its receptor, CCR2. This was followed by greater macrophage-positivity in AngII-exposed aortae. In contrast to the delayed effects on atherosclerosis, 20% of mice were found to have abdominal aneurysms at the end of AngII-exposure. This effect was not contingent on blood pressure. Moreover, despite amplification in atherosclerosis following AngII, no aneurysms were found 14 weeks later. Our studies reveal that even transient exposure to AngII primes the vessel for subsequent amplification of atherosclerosis which involves activation of MCP-1/CCR2 and influx of macrophages into the nascent atherosclerotic plaque. By contrast, transient AngII-exposure causes prompt aneurysm formation that does not parallel atherosclerosis and disappears even in the face of progressively greater atherosclerotic lesions.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Angiotensin II/metabolism
- Angiotensin II/toxicity
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/etiology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Apolipoproteins E/deficiency
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Chemokine CCL2/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Disease Models, Animal
- Female
- Follow-Up Studies
- Hyperlipidemias/complications
- Hyperlipidemias/metabolism
- Immunohistochemistry
- Infusions, Intravenous
- Mice
- Mice, Inbred C57BL
- RNA/genetics
- Receptors, CCR2
- Receptors, Chemokine/drug effects
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
Collapse
Affiliation(s)
- Nobuhiko Ayabe
- Department of Pediatrics, MCN C4204, Vanderbilt University Medical Center, Nashville, TN 37232-2584, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Grundmann S, Hoefer I, Ulusans S, van Royen N, Schirmer SH, Ozaki CK, Bode C, Piek JJ, Buschmann I. Anti-tumor necrosis factor-{alpha} therapies attenuate adaptive arteriogenesis in the rabbit. Am J Physiol Heart Circ Physiol 2005; 289:H1497-505. [PMID: 15923311 DOI: 10.1152/ajpheart.00959.2004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The specific antagonists of tumor necrosis factor-alpha (TNF-alpha), infliximab and etanercept, are established therapeutic agents for inflammatory diseases such as rheumatoid arthritis and Crohn's disease. Although the importance of TNF-alpha in chronic inflammatory diseases is well established, little is known about its implications in the cardiovascular system. Because proliferation of arteriolar connections toward functional collateral arteries (arteriogenesis) is an inflammatory-like process, we tested in vivo the hypothesis that infliximab and etanercept have antiarteriogenic actions. Sixty-three New Zealand White rabbits underwent femoral artery occlusion and received infliximab, etanercept, or vehicle according to clinical dosage regimes. After 1 wk, collateral conductance, assessed with fluorescent microspheres, revealed significant inhibition of arteriogenesis (collateral conductance): 52.4 (SD 8.1), 35.2 (SD 7.7), and 33.3 (SD 10.1) ml x min(-1) x 100 mmHg(-1) with PBS, infliximab, and etanercept, respectively (P < 0.001). High-resolution angiography showed no significant differences in number of collateral arteries, but immunohistochemical analysis demonstrated a decrease in mean collateral diameter, proliferation of vascular smooth muscle cells, and reduction of leukocyte accumulation around collateral arteries in treated groups. Infliximab and etanercept bound to infiltrating leukocytes, which are important mediators of arteriogenesis. Infliximab induced monocyte apoptosis, and neither substance affected monocyte expression of the adhesion molecule Mac-1. We demonstrated that TNF-alpha serves as a pivotal modulator of arteriogenesis, which is attenuated by treatment with TNF-alpha inhibitors. Reduction of collateral conductance is most likely due to inhibition of perivascular leukocyte infiltration and subsequent lower vascular smooth muscle cell proliferation. This is the first report showing a negative influence of TNF-alpha inhibitors on collateral artery growth.
Collapse
Affiliation(s)
- Sebastian Grundmann
- Department for Internal Medicine III (Cardiology and Angiology), University Hospital Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Tang GL, Chang DS, Sarkar R, Wang R, Messina LM. The effect of gradual or acute arterial occlusion on skeletal muscle blood flow, arteriogenesis, and inflammation in rat hindlimb ischemia. J Vasc Surg 2005; 41:312-20. [PMID: 15768015 DOI: 10.1016/j.jvs.2004.11.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Current experimental models of critical limb ischemia are based on acute ischemia rather than on chronic ischemia. Human peripheral vascular disease is largely a result of chromic ischemia. We hypothesized that a model of chronic hindlimb ischemia would develop more collateral arteries, more blood flow, and less necrosis and inflammation than would acute hindlimb ischemia. We therefore developed a rat model of chronic hindlimb ischemia and compared the effects of chronic ischemia with those of acute ischemia on hindlimb skeletal muscle. METHODS Acute or chronic ischemia was induced in 36 male Sprague-Dawley rats. Chronic ischemia caused blood flow, as measured by laser Doppler scanning and confirmed by muscle oxygen tension measurements, to gradually decrease over 1 to 2 weeks after operation. RESULTS Histologic analysis showed chronic hindlimb ischemia better preserved muscle mass and architecture and stimulated capillary angiogenesis, while lacking the muscle necrosis and inflammatory cell infiltrate seen after acute ischemia. Surprisingly, the chronic ischemia group recovered dermal blood flow more slowly and less completely than did the acute ischemia group, as measured by laser Doppler (0.66 +/- 0.02 vs 0.76 +/- 0.04, P < .05) and tissue oxygen tension (0.61 +/- 0.06 vs 0.81 +/- 0.05, P < .05) at 40 days postoperatively. Consistent with poorer blood flow recovery, chronic ischemia resulted in smaller diameter collateral arteries (average diameter of the five largest collaterals on angiogram was 0.01 +/- 0.0003 mm vs 0.013 +/- 0.0007 mm for acute, P < .005 at 40 days postoperatively). Acute ischemia resulted in decreased tissue concentrations of vascular endothelial growth factor (VEGF) (0.96 +/- 0.23 pg/mg of muscle for acute vs 4.4 +/- 0.75 and 4.8 +/- 0.75 pg/mg of muscle for unoperated and chronic, respectively, P < .05 acute vs unoperated), and in increased tissue concentrations of interleukin (IL)-1beta (7.3 +/- 4.0 pg/mg of muscle for acute vs undetectable and 1.7 +/- 1.6 pg/mg of muscle for unoperated and chronic, respectively, P < 0.05 acute vs unoperated). CONCLUSIONS We describe here the first model of chronic hindlimb ischemia in the rat. Restoration of blood flow after induction of hindlimb ischemia is dependent on the rate of arterial occlusion. This difference in blood flow recovery correlates with distinct patterns of muscle necrosis, inflammatory cell infiltration, and cytokine induction in the ischemic muscle. Differences between models of acute and chronic hindlimb ischemia may have important consequences for future studies of mechanisms regulating arteriogenesis and for therapeutic approaches aimed at promoting arteriogenesis in humans suffering from critical limb ischemia. CLINICAL RELEVANCE Despite the substantial clinical differences between acute and chronic ischemia, researchers attempting to develop molecular therapies to treat critical limb ischemia have only tested those therapies in experimental models of acute hindlimb ischemia. We present here a novel model of chronic hindlimb ischemia in the rat. We further demonstrate that when hindlimb ischemia is developed chronically, collateral artery development is poorer than when hindlimb ischemia is developed acutely. These findings suggest that further tests of molecular therapies for critical limb ischemia should be performed in chronic hindlimb ischemia models rather than in acute hindlimb ischemia models.
Collapse
Affiliation(s)
- Gale L Tang
- Pacific Vascular Research Laboratory, Department of Surgery, Division of Vascular Surgery, University of California, San Francisco 94143-0222, USA
| | | | | | | | | |
Collapse
|
75
|
da Cunha V, Tham DM, Martin-McNulty B, Deng G, Ho JJ, Wilson DW, Rutledge JC, Vergona R, Sullivan ME, Wang YXJ. Enalapril attenuates angiotensin II-induced atherosclerosis and vascular inflammation. Atherosclerosis 2005; 178:9-17. [PMID: 15585195 DOI: 10.1016/j.atherosclerosis.2004.08.023] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Revised: 07/27/2004] [Accepted: 08/10/2004] [Indexed: 01/08/2023]
Abstract
Angiotensin converting enzyme (ACE) inhibitors prevent a wide variety of key events underlying atherogenesis. Whether these actions depend solely on reduction of angiotensin II (Ang II) generation is still to be determined. This study was undertaken to determine whether enalapril, an ACE inhibitor, prevents atherosclerosis and vascular inflammation induced by Ang II in apolipoprotein E-deficient (apoE-KO) mice. Subcutaneous infusion of Ang II (1.44 mg/(kg day)) for 4 weeks increased blood pressure and accelerated atherosclerosis development in the carotid arteries. The expression of the endothelial adhesion molecules E-selectin, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), as well as the chemokines monocyte chemotactic protein-1 (MCP-1) and macrophage-colony stimulating factor (M-CSF) was up-regulated in the aortas of Ang II-treated mice. Enalapril co-treatment (25 mg/(kg day), in drinking water) prevented the development of atherosclerosis without affecting blood pressure or circulating cholesterol. In addition to preventing the Ang II-induced over-expression of adhesion molecules and chemokines in the aorta, enalapril up-regulated the expression of peroxisome proliferator-activated receptors (PPARs)-alpha and -gamma, potential anti-inflammatory transcription factors. In the aortic arch, a lesion-prone site, the co-treatment with enalapril reduced the percentage of arterial wall occupied by macrophages and foam cells, medial sclerosis and elastin reduplication. Together, these data suggest an important role for Ang II-independent mechanisms in the antiatherogenic and anti-inflammatory effects of ACE inhibitors.
Collapse
Affiliation(s)
- Valdeci da Cunha
- Department of Pharmacology, Berlex Biosciences, 2600 Hilltop Drive, PO Box 4099, Richmond, CA 94806, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Raghavendran K, Davidson BA, Mullan BA, Hutson AD, Russo TA, Manderscheid PA, Woytash JA, Holm BA, Notter RH, Knight PR. Acid and particulate-induced aspiration lung injury in mice: importance of MCP-1. Am J Physiol Lung Cell Mol Physiol 2005; 289:L134-43. [PMID: 15778247 DOI: 10.1152/ajplung.00390.2004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A model of aspiration lung injury was developed in WT C57BL/6 mice to exploit genetically modified animals on this background, i.e., MCP-1(-/-) mice. Mice were given intratracheal hydrochloric acid (ACID, pH 1.25), small nonacidified gastric particles (SNAP), or combined acid plus small gastric particles (CASP). As reported previously in rats, lung injury in WT mice was most severe for "two-hit" aspiration from CASP (40 mg/ml particulates) based on the levels of albumin, leukocytes, TNF-alpha, IL-1beta, IL-6, MCP-1, KC, and MIP-2 in bronchoalveolar lavage (BAL) at 5, 24, and 48 h. MCP-1(-/-) mice given 40 mg/ml CASP had significantly decreased survival compared with WT mice (32% vs. 80% survival at 24 h and 0% vs. 72% survival at 48 h). MCP-1(-/-) mice also had decreased survival compared with WT mice for CASP aspirates containing reduced particulate doses of 10-20 mg/ml. MCP-1(-/-) mice given 5 mg/ml CASP had survival similar to WT mice given 40 mg/ml CASP. MCP-1(-/-) mice also had differing responses from WT mice for several inflammatory mediators in BAL (KC or IL-6 depending on the particle dose of CASP and time of injury). Histopathology of WT mice with CASP (40 mg particles/ml) showed microscopic areas of compartmentalization with prominent granuloma formation by 24 h, whereas lung tissue from MCP-1(-/-) mice had severe diffuse pneumonia without granulomas. These results indicate that MCP-1 is important for survival in murine aspiration pneumonitis and appears to act partly to protect uninjured lung regions by promoting isolation and compartmentalization of tissue with active inflammation.
Collapse
Affiliation(s)
- Krishnan Raghavendran
- Department of Surgery, University at Buffalo State University of New York, 14214, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Muhs A, Lenter MC, Seidler RW, Zweigerdt R, Kirchengast M, Weser R, Ruediger M, Guth B. Nonviral monocyte chemoattractant protein-1 gene transfer improves arteriogenesis after femoral artery occlusion. Gene Ther 2005; 11:1685-93. [PMID: 15470479 DOI: 10.1038/sj.gt.3302360] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Local infusion of recombinant monocyte chemoattractant protein-1 (MCP-1) has been shown to enhance collateral artery formation in rabbit and pig hindlimb models. Owing to clinical disadvantages of protein infusion, a nonviral, liposome-based MCP-1 gene transfer was developed. Collateralization in a porcine hindlimb model served to provide a proof-of-principle for the functional benefit of MCP-1 overexpression. Development of arterial conductance as a measure of functionally relevant collateralization was evaluated in occluded as well as untreated hindlimbs in each animal. At the time of occlusion, MCP-1 and control DNA/DC-30 lipoplexes were transferred to femoral arteries of Goettingen minipigs (two therapeutic MCP-1 groups: 2 and 4 microg and one control group), using the Infiltrator local drug-delivery device. At 2 weeks following occlusion, collateralization was determined as changes in peripheral haemodynamic conductance, peripheral over aortic blood pressure ratio and angiographically visible morphology of the peripheral vessel tree. Nonviral MCP-1 gene transfer significantly improved peripheral conductance (control 11.69+/-2.78%, 2 microg 23.81+/-2.81%, P<0.05 and 4 microg 23.36+/-3.1%, P<0.05; n=12 per group) as well as the ratio of peripheral over aortic blood pressure (control 0.64+/-0.03%, 2 microg 0.75+/-0.02%, P<0.05 and 4 mug 0.75+/-0.02%, P<0.05; n=12 per group) when compared to the untreated controls 2 weeks after occlusion. Thus, it could be demonstrated for the first time that in situ overexpression of MCP-1 following local nonviral gene transfer is a potential approach to improve peripheral collateralization.
Collapse
Affiliation(s)
- A Muhs
- Cardion AG, Erkrath, Germany
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Affiliation(s)
- Timothy S Sadiq
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
79
|
Abstract
Stimulation of collateral artery growth is an attractive alternative treatment modality for patients with coronary or peripheral artery disease. Decades of basic research have led to a reasonable understanding of the mechanisms behind collateral artery growth although 'bench research' is still absolutely warranted for better understanding. It is some 7 years ago that the first clinical trials on therapeutic manipulation of the collateral circulation were published and this field is still relatively new and in large parts unexplored. Arteriogenesis, namely the growth of large collateral conductance arteries, seems to be the best biological substrate candidate for therapeutic manipulation. Future studies in this field will have to cope with problems of substance choice, clinical detection methods and unwanted side-effects.
Collapse
Affiliation(s)
- Niels van Royen
- Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
80
|
Abstract
Understanding the increasingly complex role of chemokines in various manifestations of atherosclerotic vascular disease and the apparent redundancy in their expression requires improved concepts defining the specialization and cooperation of chemokines in regulating the recruitment of mononuclear cells to vascular lesions. In an attempt to elaborate such models, this review highlights recent insights into the functional role of chemokines in mediating distinct steps during the atherogenic recruitment of monocytes and T cells obtained in genetically deficient mice and in suitable models. A particular focus is placed on the contribution of platelet chemokines deposited on endothelium for monocyte arrest, on differences in the involvement of chemokines between recruitment to native lesions and to neointimal lesions after arterial injury, and on closely related functions of macrophage migration inhibitory factor, a cytokine with considerable structural homology to chemokines. As an evolving aspect of atherosclerotic vascular disease, a role of chemokines, foremost stromal cell-derived factor-1alpha, in the recruitment of mononuclear progenitors of vascular cells during neointimal hyperplasia, endothelial recovery, and angiogenesis is discussed. The functional diversity and pleiotropy of chemokines in and beyond mononuclear cell recruitment awaits further elucidation to enable therapeutic targeting of atherogenesis by context-specific blockade of nonoverlapping chemokine receptor pairs.
Collapse
Affiliation(s)
- Christian Weber
- Kardiovaskuläre Molekularbiologie, Universitätsklinikum Aachen, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | | | | |
Collapse
|
81
|
Abstract
Our increasing appreciation of the importance of inflammation in vascular disease has focused attention on the molecules that direct the migration of leukocytes from the blood stream to the vessel wall. In this review, we summarize roles of the chemokines, a family of small secreted proteins that selectively recruit monocytes, neutrophils, and lymphocytes to sites of vascular injury, inflammation, and developing atherosclerosis. Chemokines induce chemotaxis through the activation of G-protein-coupled receptors, and the receptors that a given leukocyte expresses determines the chemokines to which it will respond. Monocyte chemoattractant protein 1 (MCP-1), acting through its receptor CCR2, appears to play an early and important role in the recruitment of monocytes to atherosclerotic lesions and in the formation of intimal hyperplasia after arterial injury. Acute thrombosis is an often fatal complication of atherosclerotic plaque rupture, and recent evidence suggests that MCP-1 contributes to thrombin generation and thrombus formation by generating tissue factor. Because of their critical roles in monocyte recruitment in vascular and nonvascular diseases, MCP-1 and CCR2 have become important therapeutic targets, and efforts are underway to develop potent and specific antagonists of these and related chemokines.
Collapse
Affiliation(s)
- Israel F Charo
- Gladstone Institute of Cardiovascular Disease, PO Box 419100, San Francisco, CA 94141-9100, USA.
| | | |
Collapse
|
82
|
Hong KH, Ryu J, Han KH. Monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth factor-A. Blood 2004; 105:1405-7. [PMID: 15498848 DOI: 10.1182/blood-2004-08-3178] [Citation(s) in RCA: 293] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) has been recognized as an angiogenic chemokine. In the present study, we investigated the detailed mechanism by which MCP-1 induces angiogenesis. We found that MCP-1 up-regulated hypoxia-inducible factor 1 alpha (HIF-1 alpha) gene expression in human aortic endothelial cells (HAECs), which induced vascular endothelial growth factor-A(165) (VEGF-A(165)) expression in the aortic wall and HAECs through activation of p42/44 mitogen-activated protein kinase (MAPK). In vivo angiogenesis assay using chick chorioallantoic membrane (CAM) showed that MCP-1-induced angiogenesis was as potent as that induced by VEGF-A(165) and completely inhibited by a VEGF inhibitor, Flt(2-11). The inhibition of RhoA small G protein did not affect MCP-1-induced VEGF-A(165) production and secretion but completely blocked both MCP-1- and VEGF-A-induced new vessel formation, as determined by CAM assay. These results suggest that MCP-1-induced angiogenesis is composed largely of 2 sequential steps: the induction of VEGF-A gene expression by MCP-1 and the subsequent VEGF-A-induced angiogenesis.
Collapse
MESH Headings
- Angiogenesis Inducing Agents/administration & dosage
- Animals
- Aorta, Abdominal/physiology
- Aorta, Thoracic/physiology
- Cells, Cultured
- Chemokine CCL2/administration & dosage
- Chemokine CCL2/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit
- Male
- Neovascularization, Physiologic/physiology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Kyung Hee Hong
- Department of Cardiology, University of Ulsan College of Medicine, Asan Medical Center, 388-1 Pungnap-2 dong Songpa-gu 138-736, Seoul, South Korea
| | | | | |
Collapse
|
83
|
Tang G, Charo DN, Wang R, Charo IF, Messina L. CCR2-/- knockout mice revascularize normally in response to severe hindlimb ischemia. J Vasc Surg 2004; 40:786-95. [PMID: 15472609 DOI: 10.1016/j.jvs.2004.07.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Monocyte chemoattractant protein-1 (MCP-1) is reported to stimulate ischemia-induced arteriogenesis (collateral artery development) by recruiting monocytes and macrophages into areas of active arteriogenesis. To determine whether the MCP-1-mediated response occurs through its receptor, CC-chemokine receptor 2 (CCR2), we induced hindlimb ischemia in mice lacking the receptor for MCP-1 (CCR2 -/- ) and measured limb blood flow recovery, collateral artery development, and monocyte and macrophage recruitment. METHODS AND RESULTS Hindlimb ischemia was induced by excising the left femoral artery in CCR2 -/- and wild-type mice. Hindlimb blood flow recovery, as measured using laser Doppler perfusion imaging, was equivalent in both groups ( P = .78 for foot and P = 0.38 for calf). Collateral artery development, as measured by angiography at postoperative day 14 and 31, likewise did not differ between the 2 groups ( P = .46 and P = .67). Counts of monocytes and macrophages in calf and thigh muscle sections of mice sacrificed on postoperative day 7 revealed that although CCR2 -/- mice recruited 44% fewer monocytes and macrophages to areas of ischemia in the calf, they recruited similar numbers of monocytes and macrophages to areas of active arteriogenesis in the thigh. Intercellular adhesion molecule-1 and MCP-1 mRNA levels were higher in the thigh muscle of CCR2 -/- mice than in wild-type mice (5.5-fold and 42.3-fold induction operated to unoperated vs 2.6-fold and 6.1-fold induction operated to unoperated, respectively). CONCLUSIONS Blood flow recovery, arteriogenesis, and monocyte and macrophage recruitment to the thigh was normal in CCR2 -/- mice. However, monocyte and macrophage recruitment to the ischemic calf was diminished in CCR2 -/- mice. Our results show that MCP-1 signaling through CCR2 is not required for physiologic arteriogenesis in response to severe hindlimb ischemia. ICAM-1 upregulation may substitute for MCP-1 signaling through CCR2 in order to allow normal arteriogenesis in CCR2 -/- mice.
Collapse
Affiliation(s)
- Gale Tang
- Pacific Vascular Research Laboratory, Department of Surgery, Division of Vascular Surgery, University of California, San Francisco 94143-0222, USA
| | | | | | | | | |
Collapse
|
84
|
Nobuhiko A, Suganuma E, Babaev VR, Fogo A, Swift LL, Linton MF, Fazio S, Ichikawa I, Kon V. Angiotensin II amplifies macrophage-driven atherosclerosis. Arterioscler Thromb Vasc Biol 2004; 24:2143-8. [PMID: 15374850 DOI: 10.1161/01.atv.0000145607.03879.e0] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We evaluated the role of angiotensin II (AII) in a marrow-derived macrophage-driven model of atherosclerosis. METHODS AND RESULTS Eight-week-old C57BL/6 wild-type mice were reconstituted with bone marrow harvested from apolipoprotein E-deficient (apoE-/---> apoE+/+) or wild-type for apoE gene (apoE+/+--> apoE+/+) mice. At 20 weeks, mice were exposed to either AII (1000 ng/kg per minute subcutaneously) or saline for 2 weeks. Animals did not differ in body weight, blood pressure, cholesterol/triglycerides, or peripheral blood monocyte counts. ApoE-/---> apoE+/+ mice exposed to AII had 3-fold greater atherosclerotic area than saline-treated apoE-/---> apoE+/+ mice. By contrast, AII did not affect atherosclerosis in apoE+/+--> apoE+/+ mice. Macrophage-positive areas were increased by AII in mice reconstituted with either apoE-deficient or apoE-competent marrow. AII also significantly increased fragmentation of elastin laminae in both apoE-/---> apoE+/+ and apoE+/+--> apoE+/+ mice. In vitro, AII caused greater increase in monocyte chemoattractant protein-1-stimulated migration of macrophages harvested from AII-infused versus saline-infused mice. CONCLUSIONS The current studies reveal that AII has both initiating and sustaining proatherogenic effects. By promoting macrophage migration into the vascular intima, AII is pivotal in initiating atherosclerosis; by promoting elastin breaks, a novel mechanism implicated in migration and proliferation of smooth muscle cells, AII may be pivotal in subsequent development and expansion of atherosclerotic lesion.
Collapse
Affiliation(s)
- Ayabe Nobuhiko
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tenn, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Ishibashi M, Egashira K, Zhao Q, Hiasa KI, Ohtani K, Ihara Y, Charo IF, Kura S, Tsuzuki T, Takeshita A, Sunagawa K. Bone marrow-derived monocyte chemoattractant protein-1 receptor CCR2 is critical in angiotensin II-induced acceleration of atherosclerosis and aneurysm formation in hypercholesterolemic mice. Arterioscler Thromb Vasc Biol 2004; 24:e174-8. [PMID: 15331433 DOI: 10.1161/01.atv.0000143384.69170.2d] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Angiotensin II (Ang II) is implicated in atherogenesis by activating inflammatory responses in arterial wall cells. Ang II accelerates the atherosclerotic process in hyperlipidemic apoE-/- mice by recruiting and activating monocytes. Monocyte chemoattractant protein-1 (MCP-1) controls monocyte-mediated inflammation through its receptor, CCR2. The roles of leukocyte-derived CCR2 in the Ang II-induced acceleration of the atherosclerotic process, however, are not known. We hypothesized that deficiency of leukocyte-derived CCR2 suppresses Ang II-induced atherosclerosis. METHODS AND RESULTS A bone marrow transplantation technique (BMT) was used to develop apoE-/- mice with and without deficiency of CCR2 in leukocytes (BMT-apoE-/-CCR2+/+ and BMT-apoE-/-CCR2-/- mice). Compared with BMT-apoE-/-CCR2+/+ mice, Ang II-induced increases in atherosclerosis plaque size and abdominal aortic aneurysm formation were suppressed in BMT-apoE-/-CCR2-/- mice. This suppression was associated with a marked decrease in monocyte-mediated inflammation and inflammatory cytokine expression. CONCLUSIONS Leukocyte-derived CCR2 is critical in Ang II-induced atherosclerosis and abdominal aneurysm formation. The present data suggest that vascular inflammation mediated by CCR2 in leukocytes is a reasonable target of therapy for treatment of atherosclerosis.
Collapse
Affiliation(s)
- Minako Ishibashi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Song J, Cottler PS, Klibanov AL, Kaul S, Price RJ. Microvascular remodeling and accelerated hyperemia blood flow restoration in arterially occluded skeletal muscle exposed to ultrasonic microbubble destruction. Am J Physiol Heart Circ Physiol 2004; 287:H2754-61. [PMID: 15319212 DOI: 10.1152/ajpheart.00144.2004] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We showed previously that microbubble destruction with pulsed 1-MHz ultrasound creates a bioeffect that stimulates arteriogenesis and a chronic increase in hyperemia blood flow in normal rat muscle. Here we tested whether ultrasonic microbubble destruction can be used to create a microvascular remodeling response that restores hyperemia blood flow to rat skeletal muscle affected by arterial occlusion. Pulsed ultrasound (1 MHz) was applied to gracilis muscles in which the lateral feed artery was occluded but the medial feed artery was left intact. Control muscles were similarly occluded but did not receive ultrasound, microbubbles, or both. Hyperemia blood flow and number of smooth muscle (SM) alpha-actin-positive vessels, >30-mum arterioles, and capillaries per fiber were determined 7, 14, and 28 days after treatment. In ultrasound-microbubble-treated muscles, lateral region hyperemia blood flow was increased at all time points and restored to normal at day 28. The number of SM alpha-actin vessels per fiber was increased over control in this region at days 7 and 14 but decreased by day 28, when larger-diameter arterioles became more prevalent in the medial region. The number of capillaries per fiber was increased over control only at day 7 in the lateral region and only at days 7 and 14 in the medial region, indicating that the angiogenesis response was transient and likely did not contribute significantly to flow restoration at day 28. We conclude that ultrasonic microbubble destruction can be tailored to stimulate an arteriogenesis response that restores hyperemia blood flow to skeletal muscle in a rat model of arterial occlusion.
Collapse
Affiliation(s)
- Ji Song
- Dept. of Biomedical Engineering, Univ. of Virginia, Box 800759, UVA Health System, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
87
|
Castro C, Campistol JM, Sancho D, Sánchez-Madrid F, Casals E, Andrés V. Rapamycin attenuates atherosclerosis induced by dietary cholesterol in apolipoprotein-deficient mice through a p27 Kip1 -independent pathway. Atherosclerosis 2004; 172:31-8. [PMID: 14709354 DOI: 10.1016/j.atherosclerosis.2003.09.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Activation of immune cells and dysregulated growth and motility of vascular smooth muscle cells contribute to neointimal lesion development during the pathogenesis of vascular obstructive disease. Inhibition of these processes by the immunosuppressant rapamycin is associated with reduced neointimal thickening in the setting of balloon angioplasty and chronic graft vessel disease (CGVD). In this study, we show that rapamycin elicits a marked reduction of aortic atherosclerosis in apolipoprotein E (apoE)-null mice fed a high-fat diet despite sustained hypercholesterolemia. This inhibitory effect of rapamycin coincided with diminished aortic expression of the positive cell cycle regulatory proteins proliferating cell nuclear antigen and cyclin-dependent kinase 2. Moreover, rapamycin prevented the normal upregulation of the proatherogenic monocyte chemoattractant protein-1 (MCP-1, CCL2) seen in the aorta of fat-fed mice. Previous studies have implicated the growth suppressor p27(Kip1) in the antiproliferative and antimigratory activities of rapamycin in vitro. However, our studies with fat-fed mice doubly deficient for p27(Kip1) and apoE disclosed an antiatherogenic effect of rapamycin comparable with that found in apoE-null mice with an intact p27(Kip1) gene. Taken together, these findings extend the therapeutic application of rapamycin from the restenosis and CGVD models to the setting of diet-induced atherosclerosis. Our results suggest that rapamycin-dependent atheroprotection occurs through a p27(Kip1)-independent pathway that involves reduced expression of positive cell cycle regulators and MCP-1 within the arterial wall.
Collapse
Affiliation(s)
- Claudia Castro
- Laboratorio de Biología Vascular, Departamento de Patología y Terapia Molecular y Celular, Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, C/Jaime Roig 11, 46010, Valencia, Spain
| | | | | | | | | | | |
Collapse
|
88
|
Maksimowicz-McKinnon K, Bhatt DL, Calabrese LH. Recent advances in vascular inflammation: C-reactive protein and other inflammatory biomarkers. Curr Opin Rheumatol 2004; 16:18-24. [PMID: 14673384 DOI: 10.1097/00002281-200401000-00005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Inflammatory vascular diseases are initiated and perpetuated by the interaction of immune cells with cells of the affected vessel wall. This is directed by a network of chemical messengers, which, in a state of vascular health, exist as balanced but opposing forces. Our understanding of this highly complex process has advanced significantly in the last several decades. The detection of vascular inflammation and monitoring of this activity have long been attempted in systemic vasculitis, and, more recently, in atherosclerosis. Markers of vascular inflammation used thus far have been of limited value; few provide both adequate sensitivity and specificity for any particular disease. New insights into the pathophysiology of vascular inflammation have identified other potential markers that may improve detection and monitoring of these conditions. RECENT FINDINGS Immunomodulatory mediators of the inflammatory cascade have been identified, and their roles are being defined. There are recent data that implicate various cytokines, proteases, adhesion molecules, and acute phase proteins as participants in the generation of vascular inflammation. CONCLUSION The pursuit of highly sensitive and specific markers of vascular inflammation has produced a wealth of information that has been instrumental in advancing our comprehension of this complex process. Further studies will establish the role of these new markers in the diagnosis, monitoring, and prognostication of inflammatory vascular disease.
Collapse
|
89
|
Tayebjee MH, Lip GYH, MacFadyen RJ. Collateralization and the response to obstruction of epicardial coronary arteries. QJM 2004; 97:259-72. [PMID: 15100419 DOI: 10.1093/qjmed/hch053] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Occlusive coronary disease is an important cause of global morbidity and mortality. While mechanical revascularization is effective, some individuals are not amenable to such interventions, and have a poorer prognosis. However, collateral circulation can protect and preserve myocardium around the time of coronary occlusion, contribute to better residual myocardial contractility, and lessen symptoms. We describe the anatomy and physiology of coronary collateralization, its component parts (angiogenesis and arteriogenesis), the current methods for definition of the collateral response and how this might be manipulated. The manipulation of this process is a realistic possibility for future adjuvant treatment of coronary artery disease.
Collapse
Affiliation(s)
- M H Tayebjee
- Haemostasis Thrombosis and Vascular Biology Unit, University Department of Medicine, City Hospital, Birmingham, UK
| | | | | |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW While it is well established that angiotensin II promotes cardiovascular and renal disorders, recent evidence has indicated a pivotal role in atherosclerotic disease which is distinguished by the central abnormality of lipid accumulation within the vascular wall. RECENT FINDINGS Studies published in the last year show that angiotensin II activity is increased in atherosclerosis, but even a transient elevation in angiotensin II potentiates the disease. The downstream hormone, aldosterone, has vasculopathic effects in conjunction with, as well as independently of, angiotensin II. The mechanism for angiotensin II injury includes potentiation of damage by known risk factors such as hypertension, hyperlipidemia, diabetes and insulin resistance, falling estrogens and inflammation. In addition, angiotensin II has direct effects on cellular proliferation, hypertrophy, apoptosis, and synthesis/degradation of matrix proteins and collagen that underlie development and progression of atherosclerosis as well as stability of the plaque. Antagonism of angiotensin II actions, therefore, offers the possibility of interfering with these direct and indirect effects and lessening the progression of atherosclerosis, stabilizing vulnerable plaques, and even reversing the disease. SUMMARY Angiotensin is increased in atherosclerosis, and increased angiotensin II amplifies atherosclerosis by modulating individual risk factors as well as by directly affecting lipid metabolism, the vascular response to lipid accumulation, and plaque stability. Antagonism of angiotensin II actions not only lessens the progression of atherosclerosis, but stabilizes the plaque and may even cause regression of the disease.
Collapse
Affiliation(s)
- Valentina Kon
- Vanderbilt University Medical Center, Nashville, Tennessee 37232-2584, USA.
| | | |
Collapse
|
91
|
van Royen N, Voskuil M, Hoefer I, Jost M, de Graaf S, Hedwig F, Andert JP, Wormhoudt TAM, Hua J, Hartmann S, Bode C, Buschmann I, Schaper W, van der Neut R, Piek JJ, Pals ST. CD44 Regulates Arteriogenesis in Mice and Is Differentially Expressed in Patients With Poor and Good Collateralization. Circulation 2004; 109:1647-52. [PMID: 15023889 DOI: 10.1161/01.cir.0000124066.35200.18] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Arteriogenesis refers to the development of collateral conductance arteries and is orchestrated by circulating monocytes, which invade growing collateral arteries and act as suppliers of cytokines and growth factors. CD44 glycoproteins are involved in leukocyte extravasation but also in the regulation of growth factor activation, stability, and signaling. Here, we explored the role of CD44 during arteriogenesis.
Methods and Results—
CD44 expression increases strongly during collateral artery growth in a murine hind-limb model of arteriogenesis. This CD44 expression is of great functional importance, because arteriogenesis is severely impaired in CD44
−/−
mice (wild-type, 54.5±14.9% versus CD44
−/−
, 24.1±9.2%,
P
<0.001). The defective arteriogenesis is accompanied by reduced leukocyte trafficking to sites of collateral artery growth (wild-type, 29±12% versus CD44
−/−
, 18±7% CD11b-positive cells/square,
P
<0.01) and reduced expression of fibroblast growth factor-2 and platelet-derived growth factor-B protein. Finally, in patients with single-vessel coronary artery disease, the maximal expression of CD44 on activated monocytes is reduced in case of impaired collateral artery formation (poor collateralization, 1764±572 versus good collateralization, 2817±1029 AU,
P
<0.05).
Conclusions—
For the first time, the pivotal role of CD44 during arteriogenesis is shown. The expression of CD44 increases during arteriogenesis, and the deficiency of CD44 severely impedes arteriogenesis. Maximal CD44 expression on isolated monocytes is decreased in patients with a poor collateralization compared with patients with a good collateralization.
Collapse
Affiliation(s)
- N van Royen
- Department of Cardiology, Room B2-114, Academic Medical Center, University of Amsterdam, Meibergdreef 9 1105 AZ, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Hoefer IE, van Royen N, Rectenwald JE, Deindl E, Hua J, Jost M, Grundmann S, Voskuil M, Ozaki CK, Piek JJ, Buschmann IR. Arteriogenesis proceeds via ICAM-1/Mac-1- mediated mechanisms. Circ Res 2004; 94:1179-85. [PMID: 15059933 DOI: 10.1161/01.res.0000126922.18222.f0] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocyte adhesion to shear stress-activated endothelium stands as an important initial step during arteriogenesis (collateral artery growth). Using multiple approaches, we tested the hypothesis that monocyte adhesion via intercellular adhesion molecule-1 (ICAM-1) and selectin interactions is essential for adaptive arteriogenesis. Forty-eight New Zealand White rabbits received either solvent, monocyte chemoattractant protein-1 (MCP-1) alone, MCP-1 plus ICAM-mab, or MCP-1 plus an IgG2a isotype control via osmotic minipumps. After 7 days, collateral conductance was evaluated: solvent 4.01 (mL/min per 100 mm Hg), MCP-1 plus ICAM-mab 8.04 (versus solvent P=NS), and MCP-1 alone 33.11 (versus solvent P<0.05). Furthermore, the right femoral arteries of ICAM-1-/-, Mac-1-/- and mice having defective selectin interactions (FT4/7-/-) as well as their corresponding controls were ligated. One week later, perfusion ratios were determined by the use of fluorescent microspheres. FT4/7-/- mice did not show any significant difference in perfusion restoration whereas ICAM-1-/- and Mac-1-/- mice had a significant reduction in arteriogenesis as compared with matching controls (FT4/7-WT 37+/-9%, FT4/7-/- 32+/-3%, P=0.31; C57BL/6J 59+/-9%, ICAM-1-/- 36+/-8%, P<0.05; Mac-1-/- 42+/-3%, P<0.05). ICAM-1/Mac-1-mediated monocyte adhesion to the endothelium of collateral arteries is an essential step for arteriogenesis, whereas this process can proceed via selectin interaction independent mechanisms. Furthermore, in vivo treatment with monoclonal antibodies against ICAM-1 totally abolishes the stimulatory effect of MCP-1 on collateral artery growth, suggesting that the mechanism of the MCP-1-induced arteriogenesis proceeds via the localization of monocytes rather than the action of the MCP-1 molecule itself.
Collapse
Affiliation(s)
- Imo E Hoefer
- Research Group for Experimental and Clinical Arteriogenesis at the Department of Cardiology, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Ryu JW, Hong KH, Maeng JH, Kim JB, Ko J, Park JY, Lee KU, Hong MK, Park SW, Kim YH, Han KH. Overexpression of uncoupling protein 2 in THP1 monocytes inhibits beta2 integrin-mediated firm adhesion and transendothelial migration. Arterioscler Thromb Vasc Biol 2004; 24:864-70. [PMID: 15016641 DOI: 10.1161/01.atv.0000125705.28058.eb] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Uncoupling protein 2 (UCP2) belongs to the mitochondrial anion carrier family and regulates production of reactive oxygen species in macrophages. Previous studies have shown that selective genetic disruption of UCP2 in bone marrow cells results in excess accumulation of monocytes/macrophages in the vascular wall of hypercholesterolemic low-density lipoprotein receptor-deficient (LDLR-/-) mice. Here we investigated whether UCP2 regulates expression of genes involved in monocyte recruitment. METHODS AND RESULTS UCP2 overexpression in THP1 monocytes, which induced a 10-fold increase in mitochondrial UCP2 protein levels, reduced steady-state level of intracellular reactive oxygen species (ROS) and H2O2-induced ROS production. THP1 monocytes with UCP2 overexpression showed lower intracellular calcium levels and less H2O2-triggered intracellular calcium mobilization, and less protein and mRNA levels of beta2 integrins, most notably CD11b. UCP2 overexpression reduced beta2 integrin-mediated firm adhesion of monocytes to either tumor necrosis factor-alpha (TNF-alpha)-stimulated human aortic endothelial cell (HAEC) monolayers or to plates coated with intercellular adhesion molecule-1, not vascular cell adhesion molecule-1. UCP2 overexpression also inhibited cell spreading and actin polymerization in monocytes treated with TNF-alpha and monocyte chemoattractant protein-1 (MCP-1), and reduced MCP-1-induced transmigration of monocytes through HAEC monolayers. CONCLUSIONS Mitochondrial UCP2 in circulating monocytes may prevent excessive accumulation of monocytes/macrophages in the arterial wall, thereby reducing atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Je-Won Ryu
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Johnson C, Sung HJ, Lessner SM, Fini ME, Galis ZS. Matrix metalloproteinase-9 is required for adequate angiogenic revascularization of ischemic tissues: potential role in capillary branching. Circ Res 2004; 94:262-8. [PMID: 14670843 PMCID: PMC6716372 DOI: 10.1161/01.res.0000111527.42357.62] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis, an essential component of a variety of physiological and pathological processes, offers attractive opportunities for therapeutic regulation. We hypothesized that matrix metalloproteinase-9 genetic deficiency (MMP-9-/-) will impair angiogenesis triggered by tissue ischemia, induced experimentally by femoral artery ligation in mice. To investigate the role of MMP-9, we performed a series of biochemical and histological analyses, including zymography, simultaneous detection of perfused capillaries, MMP-9 promoter activity, MMP-9 protein, and macrophages in MMP-9-/- and wild-type (WT) mice. We found that ischemia resulted in doubling of capillary density in WT and no change in the MMP-9-/- ischemic tissues, which translated into increased (39%) perfusion capacity only in the WT at 14 days after ligation. We also confirmed that capillaries in the MMP-9-/- presented significantly (P<0.05) less points of capillary intersections, interpreted by us as decreased branching. The combined conclusions from simultaneous localizations of MMP-9 expression, capillaries, and macrophages suggested that macrophage MMP-9 participates in capillary branching. Transplantation of WT bone marrow into the MMP-9-/-, restored capillary branching, further supporting the contribution of bone marrow-derived macrophages in supplying the necessary MMP-9. Our study indicates that angiogenesis triggered by tissue ischemia requires MMP-9, which may be involved in capillary branching, a potential novel role for this MMP that could be exploited to control angiogenesis.
Collapse
Affiliation(s)
- Chad Johnson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University School of Medicine, Atlanta, Ga, USA
| | | | | | | | | |
Collapse
|
95
|
Silvestre JS, Gojova A, Brun V, Potteaux S, Esposito B, Duriez M, Clergue M, Le Ricousse-Roussanne S, Barateau V, Merval R, Groux H, Tobelem G, Levy B, Tedgui A, Mallat Z. Transplantation of bone marrow-derived mononuclear cells in ischemic apolipoprotein E-knockout mice accelerates atherosclerosis without altering plaque composition. Circulation 2003; 108:2839-42. [PMID: 14656923 DOI: 10.1161/01.cir.0000106161.43954.df] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bone marrow-derived mononuclear cells (BM-MNCs) enhance postischemic neovascularization, and their therapeutic use is currently under clinical investigation. We evaluated the safety of BM-MNC-based therapy in the setting of atherosclerosis. METHODS AND RESULTS Apolipoprotein E (apoE)-knockout (KO) mice were divided into 4 groups: 20 nonischemic mice receiving intravenous injection of either saline (n=10) or 10(6) BM-MNCs from wild-type animals (n=10) and 20 mice with arterial femoral ligature receiving intravenous injection of either saline (n=10) or 10(6) BM-MNCs from wild-type animals (n=10) at the time of ischemia induction. Animals were monitored for 4 additional weeks. Atherosclerosis was evaluated in the aortic sinus. BM-MNC transplantation improved tissue neovascularization in ischemic hind limbs, as revealed by the 210% increase in angiography score (P<0.0001), the 33% increase in capillary density (P=0.01), and the 65% increase in tissue Doppler perfusion score (P=0.0002). Hindlimb ischemia without BM-MNC transplantation or BM-MNC transplantation without ischemia did not affect atherosclerotic plaque size. However, transplantation of 10(6) BM-MNCs into apoE-KO mice with hindlimb ischemia induced a significant 48% to 72% increase in lesion size compared with the other 3 groups (P=0.0025), despite similar total cholesterol levels. Transplantation of 10(5) BM-MNCs produced similar results, whereas transplantation of 10(6) apoE-KO-derived BM-MNCs had neither proangiogenic nor proatherogenic effects. There was no difference in plaque composition between groups. CONCLUSIONS BM-MNC therapy is unlikely to affect atherosclerotic plaque stability in the short term. However, it may promote further atherosclerotic plaque progression in an ischemic setting.
Collapse
Affiliation(s)
- Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale U541, Hôpital Lariboisière, 41, Bd de la Chapelle, 75010 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Takahashi K, Mizuarai S, Araki H, Mashiko S, Ishihara A, Kanatani A, Itadani H, Kotani H. Adiposity elevates plasma MCP-1 levels leading to the increased CD11b-positive monocytes in mice. J Biol Chem 2003; 278:46654-60. [PMID: 13129912 DOI: 10.1074/jbc.m309895200] [Citation(s) in RCA: 238] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Obesity is currently considered as an epidemic in the western world, and it represents a major risk factor for life-threatening diseases such as heart attack, stroke, diabetes, and cancer. Taking advantage of DNA microarray technology, we tried to identify the molecules explaining the relationship between obesity and vascular disorders, comparing mRNA expression of about 12,000 genes in white adipose tissue between normal, high fat diet-induced obesity (DIO) and d-Trp34 neuropeptide Y-induced obesity in mice. Expression of monocyte chemoattractant protein-1 (MCP-1) mRNA displayed a 7.2-fold increase in obese mice as compared with normal mice, leading to substantially elevated MCP-1 protein levels in adipocytes. MCP-1 levels in plasma were also increased in DIO mice, and a strong correlation between plasma MCP-1 levels and body weight was identified. We also showed that elevated MCP-1 protein levels in plasma increased the CD11b-positive monocyte/macrophage population in DIO mice. Furthermore, infusion of MCP-1 into lean mice increased the CD11b-positive monocyte population without inducing changes in body weight. Given the importance of MCP-1 in activation of monocytes and subsequent atherosclerotic development, these results suggest a novel role of adiposity in the development of vascular disorders.
Collapse
Affiliation(s)
- Kazuhiko Takahashi
- Banyu Tsukuba Research Institute in collaboration with Merck Research Laboratories, Tsukuba, Ibaraki 300-2611, Japan
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Jost MM, Ninci E, Meder B, Kempf C, Van Royen N, Hua J, Berger B, Hoefer I, Modolell M, Buschmann I. Divergent effects of GM-CSF and TGFbeta1 on bone marrow-derived macrophage arginase-1 activity, MCP-1 expression, and matrix metalloproteinase-12: a potential role during arteriogenesis. FASEB J 2003; 17:2281-3. [PMID: 14525945 DOI: 10.1096/fj.03-0071fje] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Granulocyte/macrophage-colony stimulating factor (GM-CSF) and transforming growth factor (TGF)beta1 induce arteriogenesis in a nonischemic model of femoral artery ligation. Moreover, clinical trials demonstrated an improved collateralization after injection of bone marrow cells. In the present study, the expression of arteriogenic factors in bone marrow-derived macrophages (BMDM) was measured to verify the potential of these cells to influence collateral artery growth. GM-CSF induced in BMDM the expression of monocyte chemoattractive protein (MCP)-1, matrix-metalloproteinase (MMP)-12, and arginase-1-the latter also showing a remarkable increase in activity. During in vivo induced arteriogenesis, the accumulation rate of macrophages around proliferating collaterals was significantly increased. We also show that MCP-1 is found to be mainly expressed in the media of the vessel wall, MMP-12 in macrophages of the adventitia, and arginase at both locations. This study provides for the first time a comprehensive analysis of GM-CSF/TGFbeta1-regulated arteriogenic factors in BMDM and supports the hypothesis that arteriogenesis is a multistage mechanism, including monocyte/macrophage adhesion and transmigration, pro-arteriogenic cytokine expression, degradation of connective tissue, and collagen synthesis regulation. Selective modulation of these mechanisms as well as cell-based therapies supplying arteriogenic factors in vivo point toward new strategies to influence collateral artery growth.
Collapse
Affiliation(s)
- Marco M Jost
- Research Group for Experimental and Clinical Arteriogenesis at the Department for Internal Medicine III, Albert-Ludwigs University Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Díez-Juan A, Pérez P, Aracil M, Sancho D, Bernad A, Sánchez-Madrid F, Andrés V. Selective inactivation of p27(Kip1) in hematopoietic progenitor cells increases neointimal macrophage proliferation and accelerates atherosclerosis. Blood 2003; 103:158-61. [PMID: 14504088 DOI: 10.1182/blood-2003-07-2319] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Excessive proliferation of immune cells and vascular smooth myocytes (VSMCs) contributes to atherosclerosis. We have previously shown that whole-body inactivation of the growth suppressor p27 exacerbates atherosclerosis in apolipoprotein E-null mice (apoE-/-), and this correlated with increased proliferation of arterial macrophages and VSMCs. In the present study, we postulated that targeted disruption of bone marrow (BM) p27 is sufficient to enhance arterial macrophage proliferation and atherosclerosis. To test this hypothesis, sublethally irradiated apoE-/- mice with an intact p27 gene received a BM transplant from either apoE-/- or p27-/-apoE-/- doubly deficient donor mice and challenged with a high-cholesterol diet. Compared with mice that received an apoE-/- BM transplant, reconstitution with p27-/-apoE-/- doubly deficient marrow increased the expression of proliferating cell nuclear antigen in neointimal macrophages and accelerated aortic atherosclerosis, and this correlated with augmented aortic expression of the inflammatory cytokines CCL2/MCP-1 (monocyte chemoattractant protein 1) and CCL5/RANTES (regulated on activation, normal T-cell expressed and secreted). Overall, these findings provide evidence that p27 deficiency in hematopoietic progenitor cells enhances the inflammatory/proliferative response induced by dietary cholesterol and accelerates atherosclerosis.
Collapse
Affiliation(s)
- Antonio Díez-Juan
- Department of Molecular and Cellular Pathology and Therapy, Instituto de Biomedicina de Valencia, Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
99
|
Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003; 9:702-12. [PMID: 12778169 DOI: 10.1038/nm0603-702] [Citation(s) in RCA: 1182] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Emerging evidence suggests that bone marrow-derived endothelial, hematopoietic stem and progenitor cells contribute to tissue vascularization during both embryonic and postnatal physiological processes. Recent preclinical and pioneering clinical studies have shown that introduction of bone marrow-derived endothelial and hematopoietic progenitors can restore tissue vascularization after ischemic events in limbs, retina and myocardium. Corecruitment of angiocompetent hematopoietic cells delivering specific angiogenic factors facilitates incorporation of endothelial progenitor cells (EPCs) into newly sprouting blood vessels. Identification of cellular mediators and tissue-specific chemokines, which facilitate selective recruitment of bone marrow-derived stem and progenitor cells to specific organs, will open up new avenues of research to accelerate organ vascularization and regeneration. In addition, identification of factors that promote differentiation of the progenitor cells will permit functional incorporation into neo-vessels of specific tissues while diminishing potential toxicity to other organs. In this review, we discuss the clinical potential of vascular progenitor and stem cells to restore long-lasting organ vascularization and function.
Collapse
Affiliation(s)
- Shahin Rafii
- Department of Hematology-Oncology and Genetic Medicine, Cornell University Medical College, New York, New York, 10021 USA.
| | | |
Collapse
|