51
|
Abstract
Formation of arterial vasculature, here termed arteriogenesis, is a central process in embryonic vascular development as well as in adult tissues. Although the process of capillary formation, angiogenesis, is relatively well understood, much remains to be learned about arteriogenesis. Recent discoveries point to the key role played by vascular endothelial growth factor receptor 2 in control of this process and to newly identified control circuits that dramatically influence its activity. The latter can present particularly attractive targets for a new class of therapeutic agents capable of activation of this signaling cascade in a ligand-independent manner, thereby promoting arteriogenesis in diseased tissues.
Collapse
Affiliation(s)
- Michael Simons
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| | - Anne Eichmann
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
52
|
Yang B, Cai B, Deng P, Wu X, Guan Y, Zhang B, Cai W, Schaper J, Schaper W. Nitric Oxide Increases Arterial Endotheial Permeability through Mediating VE-Cadherin Expression during Arteriogenesis. PLoS One 2015; 10:e0127931. [PMID: 26133549 PMCID: PMC4489889 DOI: 10.1371/journal.pone.0127931] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/13/2015] [Indexed: 11/18/2022] Open
Abstract
Macrophage invasion is an important event during arteriogenesis, but the underlying mechanism is still only partially understood. The present study tested the hypothesis that nitric oxide (NO) and VE-cadherin, two key mediators for vascular permeability, contribute to this event in a rat ischemic hindlimb model. In addition, the effect of NO on expression of VE-caherin and endothelial permeability was also studied in cultured HUVECs. We found that: 1) in normal arteriolar vessels (NAV), eNOS was moderately expressed in endothelial cells (EC) and iNOS was rarely detected. In contrast, in collateral vessels (CVs) induced by simple femoral artery ligation, both eNOS and iNOS were significantly upregulated (P<0.05). Induced iNOS was found mainly in smooth muscle cells, but also in other vascular cells and macrophages; 2) in NAV VE-cadherin was strongly expressed in EC. In CVs, VE-cadherin was significantly downregulated, with a discontinuous and punctate pattern. Administration of nitric oxide donor DETA NONOate (NONOate) further reduced the amounts of Ve-cadherin in CVs, whereas NO synthase inhibitor L-NAME inhibited downregulation of VE-cadherin in CVs; 3) in normal rats Evans blue extravasation (EBE) was low in the musculus gracilis, FITC-dextron leakage was not detected in the vascular wall and few macrophages were observed in perivascular space. In contrast, EBE was significantly increased in femoral artery ligation rats, FITC-dextron leakage and increased amounts of macrophages were detected in CVs, which were further enhanced by administration of NONOate, but inhibited by L-NAME supplement; 4) in vitro experiments confirmed that an increase in NO production reduced VE-cadherin expression, correlated with increases in the permeability of HUVECs. In conclusion, our data for the first time reveal the expression profile of VE-cadherin and alterations of vascular permeability in CVs, suggesting that NO-mediated VE-cadherin pathway may be one important mechanism responsible, at least in part, for macrophage invasion during arteriogenesis.
Collapse
Affiliation(s)
- Baolin Yang
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- Department of Anatomy, School of Basic Medicine, Nanchang Univ., Nanchang, 330006, Jiangxi, P.R. China
| | - Baizhen Cai
- Dept. of Intensive Care Unit, the 3rd Xiangya Hospital, Central South Univ., Changsha, 410013, Hunan, P.R. China
| | - Panyue Deng
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- * E-mail: (WC); (PD); (WS); (JS)
| | - Xiaoqiong Wu
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South Univ., Changsha, 410013, Hunan, P.R. China
| | - Yinglu Guan
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
| | - Bin Zhang
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
| | - Weijun Cai
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- * E-mail: (WC); (PD); (WS); (JS)
| | - Jutta Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
- * E-mail: (WC); (PD); (WS); (JS)
| | - Wolfgang Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
- * E-mail: (WC); (PD); (WS); (JS)
| |
Collapse
|
53
|
Sapharikas E, Lokajczyk A, Fischer AM, Boisson-Vidal C. Fucoidan Stimulates Monocyte Migration via ERK/p38 Signaling Pathways and MMP9 Secretion. Mar Drugs 2015; 13:4156-70. [PMID: 26133555 PMCID: PMC4515609 DOI: 10.3390/md13074156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 06/17/2015] [Accepted: 06/23/2015] [Indexed: 01/21/2023] Open
Abstract
Critical limb ischemia (CLI) induces the secretion of paracrine signals, leading to monocyte recruitment and thereby contributing to the initiation of angiogenesis and tissue healing. We have previously demonstrated that fucoidan, an antithrombotic polysaccharide, promotes the formation of new blood vessels in a mouse model of hindlimb ischemia. We examined the effect of fucoidan on the capacity of peripheral blood monocytes to adhere and migrate. Monocytes negatively isolated with magnetic beads from peripheral blood of healthy donors were treated with fucoidan. Fucoidan induced a 1.5-fold increase in monocyte adhesion to gelatin (p < 0.05) and a five-fold increase in chemotaxis in Boyden chambers (p < 0.05). Fucoidan also enhanced migration 2.5-fold in a transmigration assay (p < 0.05). MMP9 activity in monocyte supernatants was significantly enhanced by fucoidan (p < 0.05). Finally, Western blot analysis of fucoidan-treated monocytes showed upregulation of ERK/p38 phosphorylation. Inhibition of ERK/p38 phosphorylation abrogated fucoidan enhancement of migration (p < 0.01). Fucoidan displays striking biological effects, notably promoting monocyte adhesion and migration. These effects involve the ERK and p38 pathways, and increased MMP9 activity. Fucoidan could improve critical limb ischemia by promoting monocyte recruitment.
Collapse
Affiliation(s)
- Elene Sapharikas
- Inserm UMR_S 1140, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris Cité, 4 Avenue de l'observatoire Paris 75006, France.
| | - Anna Lokajczyk
- Inserm UMR_S 1140, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris Cité, 4 Avenue de l'observatoire Paris 75006, France.
| | - Anne-Marie Fischer
- Inserm UMR-S 970, AP-HP, Hôpital Européen Georges Pompidou, 20 rue Leblanc Paris 75015, France.
| | - Catherine Boisson-Vidal
- Inserm UMR_S 1140, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris Cité, 4 Avenue de l'observatoire Paris 75006, France.
| |
Collapse
|
54
|
Schirmer SH, Millenaar DN, Werner C, Schuh L, Degen A, Bettink SI, Lipp P, van Rooijen N, Meyer T, Böhm M, Laufs U. Exercise promotes collateral artery growth mediated by monocytic nitric oxide. Arterioscler Thromb Vasc Biol 2015; 35:1862-71. [PMID: 26088573 DOI: 10.1161/atvbaha.115.305806] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 06/04/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Collateral artery growth (arteriogenesis) is an important adaptive response to hampered arterial perfusion. It is unknown whether preventive physical exercise before limb ischemia can improve arteriogenesis and modulate mononuclear cell function. This study aimed at investigating the effects of endurance exercise before arterial occlusion on MNC function and collateral artery growth. APPROACH AND RESULTS After 3 weeks of voluntary treadmill exercise, ligation of the right femoral artery was performed in mice. Hindlimb perfusion immediately after surgery did not differ from sedentary mice. However, previous exercise improved perfusion restoration ≤7 days after femoral artery ligation, also when exercise was stopped at ligation. This was accompanied by an accumulation of peri-collateral macrophages and increased expression of endothelial nitric oxide synthase and inducible nitric oxide synthase (iNOS) in hindlimb collateral and in MNC of blood and spleen. Systemic monocyte and macrophage depletion by liposomal clodronate but not splenectomy attenuated exercise-induced perfusion restoration, collateral artery growth, peri-collateral macrophage accumulation, and upregulation of iNOS. iNOS-deficient mice did not show exercise-induced perfusion restoration. Transplantation of bone marrow-derived MNC from iNOS-deficient mice into wild-type animals inhibited exercise-induced collateral artery growth. In contrast to sedentary controls, thrice weekly aerobic exercise training for 6 months in humans increased peripheral blood MNC iNOS expression. CONCLUSIONS Circulating mononuclear cell-derived inducible nitric oxide is an important mediator of exercise-induced collateral artery growth.
Collapse
Affiliation(s)
- Stephan H Schirmer
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.).
| | - Dominic N Millenaar
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Christian Werner
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Lisa Schuh
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Achim Degen
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Stephanie I Bettink
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Peter Lipp
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Nico van Rooijen
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Tim Meyer
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Michael Böhm
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| | - Ulrich Laufs
- From the Klinik für Innere Medizin III (S.H.S., D.N.M., C.W., L.S., A.D., S.I.B., M.B., U.L.) and Institut für Molekulare Zellbiologie (P.L.), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany; Department of Molecular Cell Biology, Faculty of Medicine, Vrije Universiteit, VUMC, Amsterdam, The Netherlands (N.R.); and Institut für Sport- und Präventivmedizin, Universität des Saarlandes, Saarbrücken, Germany (T.M.)
| |
Collapse
|
55
|
XU YIGUAN, TAN XUERUI, WANG DONGMING, WANG WEI, LI YUGUANG, WU MIN, CHEN SONGMING, WU YINGE, TAN CHUNJIANG. Elevated survivin expression in peripheral blood mononuclear cells is central to collateral formation in coronary chronic total occlusion. Int J Mol Med 2015; 35:1501-10. [PMID: 25816072 PMCID: PMC4432932 DOI: 10.3892/ijmm.2015.2154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 03/09/2015] [Indexed: 02/05/2023] Open
Abstract
Survivin is essential to angiogenesis and revascularization, but its role in coronary collateral formation remains unclear. The role of survivin in peripheral blood mononuclear cells (PBMCs) of coronary chronic total occlusion (CTO) patients was investigated. Coronary CTO patients (n=46; mean age 60.1±8.5, male 54.3%) (CTO group) and normal control patients (n=18; mean age 58.0±10.0, male 55.6%) underwent angiographic collateral vessel grading by Rentrop classification (C0 - C3) and provided peripheral blood between June 2006 and February 2007. Rat hind limb ischemia models were constructed using four equal groups of Sprague-Dawley rats (n=36): normal control, sham operation, operation and granulocyte macrophage colony-stimulating factor (GM-CSF). PBMC numbers and characteristics, collateral vessels, survivin, CD4, CD8, CD44, vascular endothelial growth factor (VEGF) and intercellular adhesion molecule-1 (ICAM-1) expression were determined using RT-PCR, flow cytometry, immunocytochemistry and western blot analysis. PBMC survivin mRNA and protein expression levels were higher in patients with good collateral circulation (C2 + C3) than in patients with no collateral flow (C0) (all P<0.05). Survivin single-positive and survivin and CD8, VEGF and ICAM-1 double-positive percentages were elevated in patients with good collateral circulation compared to those with normal and no collateral flow (all P<0.05), consistent with the rat model results, wherein higher survivin levels produced significantly larger and more visible collateral vessels. In conclusion, elevated survivin expression in PBMCs, particularly survivin and CD8, VEGF, and ICAM-1 double-positive PBMCs, may be crucial for good collateral formation in patients with coronary CTO, as confirmed by assessment of a rat model.
Collapse
Affiliation(s)
| | | | | | | | - YUGUANG LI
- Correspondence to: Dr Yuguang Li, Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, Guangdong 515041, P.R. China, E-mail:
| | | | | | | | | |
Collapse
|
56
|
Hakimzadeh N, Verberne HJ, Siebes M, Piek JJ. The future of collateral artery research. Curr Cardiol Rev 2015; 10:73-86. [PMID: 23638829 PMCID: PMC3968596 DOI: 10.2174/1573403x113099990001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 08/13/2013] [Accepted: 09/20/2013] [Indexed: 12/20/2022] Open
Abstract
In the event of obstructive coronary artery disease, collateral arteries have been deemed an alternative blood
source to preserve myocardial tissue perfusion and function. Monocytes play an important role in modulating this process,
by local secretion of growth factors and extracellular matrix degrading enzymes. Extensive efforts have focused on developing
compounds for augmenting the growth of collateral vessels (arteriogenesis). Nonetheless, clinical trials investigating
the therapeutic potential of these compounds resulted in disappointing outcomes. Previous studies focused on developing
compounds that stimulated collateral vessel growth by enhancing monocyte survival and activity. The limited success
of these compounds in clinical studies, led to a paradigm shift in arteriogenesis research. Recent studies have shown genetic
heterogeneity between CAD patients with sufficient and insufficient collateral vessels. The genetic predispositions in
patients with poorly developed collateral vessels include overexpression of arteriogenesis inhibiting signaling pathways.
New directions of arteriogenesis research focus on attempting to block such inhibitory pathways to ultimately promote arteriogenesis.
Methods to detect collateral vessel growth are also critical in realizing the therapeutic potential of newly developed
compounds. Traditional invasive measurements of intracoronary derived collateral flow index remain the gold
standard in quantifying functional capacity of collateral vessels. However, advancements made in hybrid diagnostic imaging
modalities will also prove to be advantageous in detecting the effects of pro-arteriogenic compounds.
Collapse
Affiliation(s)
| | | | | | - Jan J Piek
- Department of Cardiology, Room B2-250, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
57
|
Hollander MR, Horrevoets AJG, van Royen N. Cellular and pharmacological targets to induce coronary arteriogenesis. Curr Cardiol Rev 2015; 10:29-37. [PMID: 23638831 PMCID: PMC3968592 DOI: 10.2174/1573403x113099990003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 02/28/2013] [Accepted: 04/19/2013] [Indexed: 12/21/2022] Open
Abstract
The formation of collateral vessels (arteriogenesis) to sustain perfusion in ischemic tissue is native to the body and can compensate for coronary stenosis. However, arteriogenesis is a complex process and is dependent on many different factors. Although animal studies on collateral formation and stimulation show promising data, clinical trials have failed to replicate these results. Further research to the exact mechanisms is needed in order to develop a pharmalogical stimulant. This review gives an overview of recent data in the field of arteriogenesis.
Collapse
Affiliation(s)
| | | | - Niels van Royen
- VU University Medical Center, Department of Cardiology, Room 4D-36, de Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
58
|
Schmidt-Bleek K, Kwee BJ, Mooney DJ, Duda GN. Boon and Bane of Inflammation in Bone Tissue Regeneration and Its Link with Angiogenesis. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:354-64. [PMID: 25742724 DOI: 10.1089/ten.teb.2014.0677] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Delayed healing or nonhealing of bone is an important clinical concern. Although bone, one of the two tissues with scar-free healing capacity, heals in most cases, healing is delayed in more than 10% of clinical cases. Treatment of such delayed healing condition is often painful, risky, time consuming, and expensive. Tissue healing is a multistage regenerative process involving complex and well-orchestrated steps, which are initiated in response to injury. At best, these steps lead to scar-free tissue formation. At the onset of healing, during the inflammatory phase, stationary and attracted macrophages and other immune cells at the fracture site release cytokines in response to injury. This initial reaction to injury is followed by the recruitment, proliferation, and differentiation of mesenchymal stromal cells, synthesis of extracellular matrix proteins, angiogenesis, and finally tissue remodeling. Failure to heal is often associated with poor revascularization. Since blood vessels mediate the transport of circulating cells, oxygen, nutrients, and waste products, they appear essential for successful healing. The strategy of endogenous regeneration in a tissue such as bone is interesting to analyze since it may represent a blueprint of successful tissue formation. This review highlights the interdependency of the time cascades of inflammation, angiogenesis, and tissue regeneration. A better understanding of these inter-relations is mandatory to early identify patients at risk as well as to overcome critical clinical conditions that limit healing. Instead of purely tolerating the inflammatory phase, modulations of inflammation (immunomodulation) might represent a valid therapeutic strategy to enhance angiogenesis and foster later phases of tissue regeneration.
Collapse
Affiliation(s)
- Katharina Schmidt-Bleek
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Brian J Kwee
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - David J Mooney
- 3 The Mooney Lab: Laboratory for Cell and Tissue Engineering, Harvard-School of Engineering and Applied Sciences , Cambridge, Massachusetts
| | - Georg N Duda
- 1 Julius Wolff Institut and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin , Berlin, Germany .,2 Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
59
|
Xiang L, Varshney R, Rashdan NA, Shaw JH, Lloyd PG. Placenta growth factor and vascular endothelial growth factor a have differential, cell-type specific patterns of expression in vascular cells. Microcirculation 2015; 21:368-79. [PMID: 24410720 DOI: 10.1111/micc.12113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 01/07/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE PLGF, a VEGF-A related protein, mediates collateral enlargement via monocytes but plays little role in capillary proliferation. In contrast, VEGF-A mediates both collateral enlargement and capillary proliferation. PLGF has been less thoroughly studied than VEGF-A, and questions remain regarding its regulation and function. Therefore, our goal was to characterize the expression of PLGF by vascular cells. We hypothesized that vascular SMC would express more PLGF than EC, since VEGF-A is primarily expressed by non-EC. METHODS We compared PLGF and VEGF-A across eight EC and SMC lines, then knocked down PLGF and evaluated cell function. We also assessed the effect of hypoxia on PLGF expression and promoter activity. RESULTS PLGF was most highly expressed in EC, whereas VEGF-A was most highly expressed in SMC. PLGF knockdown did not affect EC number, migration, or tube formation, but reduced monocyte migration toward EC. Monocyte migration was rescued by exogenous PLGF. Hypoxia increased PLGF protein without activating PLGF gene transcription. CONCLUSIONS PLGF and VEGF-A have distinct patterns of expression in vascular cells. EC derived PLGF may function primarily in communication between EC and circulating cells. Hypoxia increases EC PLGF expression posttranscriptionally.
Collapse
Affiliation(s)
- Lingjin Xiang
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, USA
| | | | | | | | | |
Collapse
|
60
|
Nishijima Y, Akamatsu Y, Weinstein PR, Liu J. Collaterals: Implications in cerebral ischemic diseases and therapeutic interventions. Brain Res 2015; 1623:18-29. [PMID: 25770816 DOI: 10.1016/j.brainres.2015.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/27/2015] [Accepted: 03/03/2015] [Indexed: 02/09/2023]
Abstract
Despite the tremendous progress made in the treatment of cerebrovascular occlusive diseases, many patients suffering from ischemic brain injury still experience dismal outcomes. Although rehabilitation contributes to post-stroke functional recovery, there is no doubt that interventions that promote the restoration of blood supply are proven to minimize ischemic injury and improve recovery. In response to the acutely decreased blood perfusion during arterial occlusion, arteriogenesis, the compensation of blood flow through the collateral circulation during arterial obstructive diseases can act not only in a timely fashion but also much more efficiently compared to angiogenesis, the sprouting of new capillaries, and a mechanism occurring in a delayed fashion while increases the total resistance of the vascular bed of the affected territory. Interestingly, despite the vast differences between the two vascular remodeling mechanisms, some crucial growth factors and cytokines involved in angiogenesis are also required for arteriogenesis. Understanding the mechanisms underlying vascular remodeling after ischemic brain injury is a critical step towards the development of effective therapies for ischemic stroke. The present article will discuss our current views in vascular remodeling acutely after brain ischemia, namely arteriogenesis, and some relevant clinical therapies available on the horizon in augmenting collateral flow that hold promise in treating ischemic brain injury. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yasuo Nishijima
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurosurgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yosuke Akamatsu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurosurgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Phillip R Weinstein
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA.
| |
Collapse
|
61
|
De Angelis B, Gentile P, Orlandi F, Bocchini I, Di Pasquali C, Agovino A, Gizzi C, Patrizi F, Scioli MG, Orlandi A, Cervelli V. Limb rescue: a new autologous-peripheral blood mononuclear cells technology in critical limb ischemia and chronic ulcers. Tissue Eng Part C Methods 2015; 21:423-35. [PMID: 25341088 DOI: 10.1089/ten.tec.2014.0245] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Therapeutic angiogenesis by autologous-peripheral blood mononuclear cells (A-PBMNC) implantation has been shown to be a safe and effective treatment for critical limb ischemia (CLI). We herein report our investigation of the long-term efficacy of implantation of A-PBMNC produced by selective filtration to treat patients with CLI, for which surgical bypass and/or percutaneous transluminal angioplasty are not possible. MATERIALS AND METHODS This is a prospective, and not a randomized, study based on a treated group who did not respond to conventional therapy (n=43) when implanted with A-PBMNC cells versus a historically matched control group. Patients of both groups were suffering from CLI Fontaine scale IV with chronic ulcers and various accompanying conditions (diabetes, heart disease, kidney failure, etc.). Treated patients were implanted with 12 mL of A-PBMNC, 0.2-0.3 mL for each bolus, collected by selective filtration from 120 mL of peripheral blood in the ischemic area of the limbs. Patients were not mobilized by granulocyte colony-stimulating factor, and the A-PBMNC treatment was repeated for a maximum of three times. RESULTS The A-PBMNC-treated group showed a statistically significant improvement of limb rescue of 95.3% versus 52.2% of the control group (p<0.001), and the result had been maintained for 2 years. The A-PBMNC group also showed reduction in pain at rest, increased maximum walking distance, and healing of the wound, which led to an overall improvement in the quality of life. Post-treatment radiological studies showed an improvement of vascularization with the formation of new collateral and by histological findings. Within 2 years of follow-up, none of the patients whom we treated showed any major or systemic adverse effects. CONCLUSION The local injection of A-PBMNC showed striking early and long-term effects together with a favorable safety profile, significantly decreasing the risk of amputation. Our results are comparable with published data obtained by injection of bone marrow mononuclear cells, but with a lot less invasive approach. Moreover the intraoperative selective filtration system we used is fast, safe, not operator dependent, and easy to use in a sterile operating theatre. This system aims to produce fresh A-PBMNC as a valuable treatment option, particularly for those difficult patients who cannot undergo revascularization.
Collapse
Affiliation(s)
- Barbara De Angelis
- 1 Department of Plastic and Reconstructive Surgery, University of Rome Tor Vergata , Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Söğüt E, Kadı H, Karayakalı M, Mertoğlu C. The association of plasma vitamin A and E levels with coronary collateral circulation. Atherosclerosis 2015; 239:547-51. [PMID: 25728388 DOI: 10.1016/j.atherosclerosis.2015.02.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/15/2015] [Accepted: 02/14/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To investigate if plasma levels of vitamin A and E have an association with coronary collateral development. METHODS A total of 189 patients who underwent coronary angiography and had total occlusion in at least one major epicardial coronary artery were enrolled in the study. To classify coronary collateral circulation (CCC), the Rentrop scoring system was used. Patients were classified as having poor CCC (Rentrop grades 0-1) or good CCC (Rentrop grades 2-3), and all patients were also screened for hypertension, hypercholesterolemia, diabetes, and smoking history. RESULTS There were no differences in plasma vitamin A and E levels between the two groups (vitamin A: 2.37 ± 0.65 vs. 2.35 ± 0.78, p = 0.253; vitamin E: 47.1 ± 12.8 vs. 44.6 ± 15.1, p = 0.082), and plasma vitamin A and E levels were not associated with CCC. Serum high-sensitivity C-reactive protein (hs-CRP) levels were significantly higher in patients with poor CCC (4.68 ± 2.52 vs. 3.89 ± 1.78, p = 0.001). The higher frequency of diabetes and higher serum hs-CRP levels were found to be an independent predictor for poor CCC (odds ratio = 2.44, p = 0.006; odds ratio = 1.24, p = 0.007, respectively). And a higher frequency of total occluded RCA was found to be a positive predictor for good CCC (odds ratio = 2.36, p = 0.06) in a multivariate logistic regression analysis. CONCLUSIONS We found that serum hs-CRP levels, presence of diabetes, and total occlusion of RCA have an effect on coronary collateral development. We found no correlation between plasma vitamin A and E levels and CCC.
Collapse
Affiliation(s)
- Erkan Söğüt
- Izmir Kâtip Çelebi University, Faculty of Medicine, Department of Biochemistry, Izmir, Turkey.
| | - Hasan Kadı
- Gaziosmanpaşa University, Faculty of Medicine, Department of Cardiology, Tokat, Turkey
| | - Metin Karayakalı
- Gaziosmanpaşa University, Faculty of Medicine, Department of Cardiology, Tokat, Turkey
| | - Cuma Mertoğlu
- Gaziosmanpaşa University, Faculty of Medicine, Department of Biochemistry, Tokat, Turkey
| |
Collapse
|
63
|
Yetkin E, Topal E, Erguzel N, Senen K, Heper G, Waltenberger J. Diabetes mellitus and female gender are the strongest predictors of poor collateral vessel development in patients with severe coronary artery stenosis. Angiogenesis 2015; 18:201-7. [DOI: 10.1007/s10456-015-9460-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/17/2015] [Indexed: 11/29/2022]
|
64
|
Schutt RC, Trachtenberg BH, Cooke JP, Traverse JH, Henry TD, Pepine CJ, Willerson JT, Perin EC, Ellis SG, Zhao DXM, Bhatnagar A, Johnstone BH, Lai D, Resende M, Ebert RF, Wu JC, Sayre SL, Orozco A, Zierold C, Simari RD, Moyé L, Cogle CR, Taylor DA. Bone marrow characteristics associated with changes in infarct size after STEMI: a biorepository evaluation from the CCTRN TIME trial. Circ Res 2015; 116:99-107. [PMID: 25406300 PMCID: PMC4282599 DOI: 10.1161/circresaha.116.304710] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022]
Abstract
RATIONALE Despite significant interest in bone marrow mononuclear cell (BMC) therapy for ischemic heart disease, current techniques have resulted in only modest benefits. However, selected patients have shown improvements after autologous BMC therapy, but the contributing factors are unclear. OBJECTIVE The purpose of this study was to identify BMC characteristics associated with a reduction in infarct size after ST-segment-elevation-myocardial infarction. METHODS AND RESULTS This prospective study comprised patients consecutively enrolled in the CCTRN TIME (Cardiovascular Cell Therapy Research Network Timing in Myocardial Infarction Evaluation) trial who agreed to have their BMCs stored and analyzed at the CCTRN Biorepository. Change in infarct size between baseline (3 days after percutaneous coronary intervention) and 6-month follow-up was measured by cardiac MRI. Infarct-size measurements and BMC phenotype and function data were obtained for 101 patients (mean age, 56.5 years; mean screening ejection fraction, 37%; mean baseline cardiac MRI ejection fraction, 45%). At 6 months, 75 patients (74.3%) showed a reduction in infarct size (mean change, -21.0±17.6%). Multiple regression analysis indicated that infarct size reduction was greater in patients who had a larger percentage of CD31(+) BMCs (P=0.046) and in those with faster BMC growth rates in colony-forming unit Hill and endothelial-colony forming cell functional assays (P=0.033 and P=0.032, respectively). CONCLUSIONS This study identified BMC characteristics associated with a better clinical outcome in patients with segment-elevation-myocardial infarction and highlighted the importance of endothelial precursor activity in regenerating infarcted myocardium. Furthermore, it suggests that for these patients with segment-elevation-myocardial infarction, myocardial repair was more dependent on baseline BMC characteristics than on whether the patient underwent intracoronary BMC transplantation. CLINICAL TRIAL REGISTRATION INFORMATION URL http://www.clinicaltrials.gov. Unique identifier: NCT00684021.
Collapse
Affiliation(s)
- Robert C Schutt
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Barry H Trachtenberg
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - John P Cooke
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Jay H Traverse
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Timothy D Henry
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Carl J Pepine
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - James T Willerson
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Emerson C Perin
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Stephen G Ellis
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - David X M Zhao
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Aruni Bhatnagar
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Brian H Johnstone
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Dejian Lai
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Micheline Resende
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Ray F Ebert
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Joseph C Wu
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Shelly L Sayre
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Aaron Orozco
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Claudia Zierold
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Robert D Simari
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Lem Moyé
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.).
| | - Christopher R Cogle
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| | - Doris A Taylor
- From the Houston Methodist DeBakey Heart and Vascular Center (R.C.S., B.H.T., J.P.C.) and Houston Methodist Research Institute (R.C.S., B.H.T., J.P.C.), TX; Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.); Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.); University of Florida College of Medicine, Gainesville (C.J.P., C.R.C.); Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., M.R., A.O., D.A.T.); University of Minnesota School of Medicine, Minneapolis (C.Z.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest, School of Medicine, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (A.B.); Indiana University School of Medicine, Indianapolis (B.H.J.); The University of Texas Health Science Center, School of Public Health, Houston (D.L., S.L.S., L.M.); National Heart, Lung, and Blood Institute, Bethesda, MD (R.F.E.); Stanford University, School of Medicine, CA (J.C.W.); and Kansas University Medical Center, School of Medicine, Kansas City (R.D.S.)
| |
Collapse
|
65
|
Chandraratne S, von Bruehl ML, Pagel JI, Stark K, Kleinert E, Konrad I, Farschtschi S, Coletti R, Gärtner F, Chillo O, Legate KR, Lorenz M, Rutkowski S, Caballero-Martinez A, Starke R, Tirniceriu A, Pauleikhoff L, Fischer S, Assmann G, Mueller-Hoecker J, Ware J, Nieswandt B, Schaper W, Schulz C, Deindl E, Massberg S. Critical role of platelet glycoprotein ibα in arterial remodeling. Arterioscler Thromb Vasc Biol 2014; 35:589-97. [PMID: 25550202 DOI: 10.1161/atvbaha.114.304447] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Arteriogenesis is strongly dependent on the recruitment of leukocytes, especially monocytes, into the perivascular space of growing collateral vessels. On the basis of previous findings that platelets are central players in inflammatory processes and mediate the recruitment of leukocytes, the aim of this study was to assess the role of platelets in a model of arterial remodeling. APPROACH AND RESULTS C57Bl6 wild-type mice, IL4-R/Iba mice lacking the extracellular domain of the glycoprotein Ibα (GPIbα) receptor, and mice treated with antibodies to block GPIbα or deplete circulating platelets were studied in peripheral arteriogenesis. Using a novel model of intravital 2-photon and epifluorescence imaging, we visualized and quantified the interaction of platelets with leukocytes and the vascular endothelium in vivo. We found that transient platelet adhesion to the endothelium of collateral vessels was a major event during arteriogenesis and depended on GPIbα. Furthermore, leukocyte recruitment was obviously affected in animals with defective platelet GPIbα function. In IL4-R/Iba mice, transient and firm leukocyte adhesion to the endothelium of collateral vessels, as well as leukocyte accumulation in the perivascular space, were significantly reduced. Furthermore, we detected platelet-leukocyte aggregates within the circulation, which were significantly reduced in IL4-R/Iba animals. Finally, platelet depletion and loss of GPIbα function resulted in poor reperfusion recovery as determined by laser Doppler imaging. CONCLUSIONS Thus, GPIbα-mediated interactions between platelets and endothelial cells, as well as leukocytes, support innate immune cell recruitment and promote arteriogenesis-establishing platelets as critical players in this process.
Collapse
Affiliation(s)
- Sue Chandraratne
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Marie-Luise von Bruehl
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Judith-Irina Pagel
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Konstantin Stark
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Eike Kleinert
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Ildiko Konrad
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Said Farschtschi
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Raffaele Coletti
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Florian Gärtner
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Omari Chillo
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Kyle R Legate
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Michael Lorenz
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Simon Rutkowski
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Amelia Caballero-Martinez
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Richard Starke
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Anca Tirniceriu
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Laurenz Pauleikhoff
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Silvia Fischer
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Gerald Assmann
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Josef Mueller-Hoecker
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Jerry Ware
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Bernhard Nieswandt
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Wolfgang Schaper
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Christian Schulz
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Elisabeth Deindl
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.)
| | - Steffen Massberg
- From the Medizinische Klinik und Poliklinik I, Department of Cardiology (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M.), Walter-Brendel-Centre of Experimental Medicine (S.C., M.-L.v.B., J.-I.P., K.S., E.K., I.K., S.F., R.C., F.G., O.C., K.R.L., M.L., S.R., A.C.-M., A.T., L.P., C.S., E.D., S.M.), Department of Anaesthesiology (J.-I.P.), Department of Applied Physics (K.R.L.), and Institute of Pathology (G.A., J.M.-H.), Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany (S.C., M.-L.v.B., K.S., I.K., S.F., R.C., F.G., K.R.L., M.L., A.T., C.S., S.M); Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany (S.F.); Vascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Hammersmith Campus, Imperial College London, South Kensington Campus, London, United Kingdom (R.S.); Department of Physiology and Biophysics, University of Arkansas for Medical Science, Little Rock (J.W.); Rudolf Virchow Center and DFG Research Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany (B.N.); and Max Planck Institute for Heart and Lung Research, Giessen, Germany (W.S.).
| |
Collapse
|
66
|
Desposito D, Potier L, Chollet C, Gobeil F, Roussel R, Alhenc-Gelas F, Bouby N, Waeckel L. Kinin receptor agonism restores hindlimb postischemic neovascularization capacity in diabetic mice. J Pharmacol Exp Ther 2014; 352:218-26. [PMID: 25398240 DOI: 10.1124/jpet.114.219196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Limb ischemia is a major complication of thromboembolic diseases. Diabetes worsens prognosis by impairing neovascularization. Genetic or pharmacological inactivation of the kallikrein-kinin system aggravates limb ischemia in nondiabetic animals, whereas angiotensin I-converting enzyme/kininase II inhibition improves outcome. The role of kinins in limb ischemia in the setting of diabetes is not documented. We assessed whether selective activation of kinin receptors by pharmacological agonists can influence neovascularization in diabetic mice with limb ischemia and have a therapeutic effect. Selective pseudopeptide kinin B1 or B2 receptor agonists resistant to peptidase action were administered by osmotic minipumps at a nonhypotensive dosage for 14 days after unilateral femoral artery ligation in mice previously rendered diabetic by streptozotocin. Comparison was made with ligatured, nonagonist-treated nondiabetic and diabetic mice. Diabetes reduced neovascularization, assessed by microangiography and histologic capillary density analysis, by roughly 40%. B1 receptor agonist or B2 receptor agonist similarly restored neovascularization in diabetic mice. Neovascularization in agonist-treated diabetic mice was indistinguishable from nondiabetic mice. Both treatments restored blood flow in the ischemic hindfoot, measured by laser-Doppler perfusion imaging. Macrophage infiltration increased 3-fold in the ischemic gastrocnemius muscle during B1 receptor agonist or B2 receptor agonist treatment, and vascular endothelial growth factor (VEGF) level increased 2-fold. Both treatments increased, by 50-100%, circulating CD45/CD11b-positive monocytes and CD34(+)/VEGFR2(+) progenitor cells. Thus, selective pharmacological activation of B1 or B2 kinin receptor overcomes the effect of diabetes on postischemic neovascularization and restores tissue perfusion through monocyte/macrophage mobilization. Kinin receptors are potential therapeutic targets in limb ischemia in diabetes.
Collapse
Affiliation(s)
- Dorinne Desposito
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Louis Potier
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Catherine Chollet
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Fernand Gobeil
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ronan Roussel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Francois Alhenc-Gelas
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Nadine Bouby
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ludovic Waeckel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| |
Collapse
|
67
|
Kwee BJ, Mooney DJ. Manipulating the intersection of angiogenesis and inflammation. Ann Biomed Eng 2014; 43:628-40. [PMID: 25316589 DOI: 10.1007/s10439-014-1145-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/27/2014] [Indexed: 12/18/2022]
Abstract
There exists a critical need to develop strategies that promote blood vessel formation (neovascularization) in virtually all tissue engineering and regenerative medicine efforts. While research typically focuses on understanding and exploiting the role of angiogenic factors and vascular cells on new blood vessel formation, the activity of the immune system is being increasingly recognized to impact vascular formation and adaptation. This review will provide both an overview of the intersection of angiogenesis and the immune system, and how biomaterials may be designed to promote favorable interactions between these two systems to promote effective vascularization.
Collapse
Affiliation(s)
- Brian J Kwee
- School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 319, Cambridge, MA, 02138, USA
| | | |
Collapse
|
68
|
Tang GL, Weitz K. Impaired arteriogenesis in syndecan-1(-/-) mice. J Surg Res 2014; 193:22-7. [PMID: 25194470 DOI: 10.1016/j.jss.2014.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/26/2014] [Accepted: 08/01/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Collateral artery development (arteriogenesis) is an important compensatory response to arterial occlusion caused by atherosclerosis. The heparan sulfate proteoglycan syndecan-1 (sdc1) has previously been shown to affect the response to arterial injury but has yet been studied in arteriogenesis. We tested the hypothesis that sdc1 knockout (sdc1(-/-)) mice would revascularize more poorly than wild type (wt) mice, and then used bone marrow transplantation experiments to determine whether sdc1's effect on arteriogenesis was due to its presence in the local tissue environment or in bone marrow derived cells. MATERIALS AND METHODS Hindlimb ischemia was induced by femoral artery ligation in wt and sdc1(-/-) female mice as well as in wt and sdc1(-/-) female mice transplanted with wt bone marrow or in wt mice transplanted with sdc1(-/-) bone marrow. Blood flow recovery was assessed by laser Doppler perfusion imaging. Arteriogenesis was assessed by measuring the diameter of the dominant collateral pathway after pressure perfusion fixation and intra-aortic contrast injection at 28 d. Immunohistochemistry was used to assess angiogenesis and peri-collateral macrophage infiltration at 7 d, postoperatively. RESULTS Sdc1(-/-) mice had impaired blood flow recovery in response to hindlimb ischemia. This impaired recovery was not secondary to a defect in capillary angiogenesis nor was it due to decreased peri-collateral macrophage infiltration. Wt bone marrow did not rescue the impaired recovery of sdc1(-/-) mice. CONCLUSIONS Sdc1 affects arteriogenesis in response to hindlimb ischemia and is required in the local tissue environment for normal arteriogenesis.
Collapse
Affiliation(s)
- Gale L Tang
- VA Puget Sound Health Care System, University of Washington, Seattle, Washington; Division of Vascular Surgery, Department of Surgery, University of Washington, Seattle, Washington.
| | - Kevin Weitz
- Division of Vascular Surgery, Department of Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
69
|
Kumar AHS, Martin K, Doyle B, Huang CL, Pillai GKM, Ali MT, Skelding KA, Wang S, Gleeson BM, Jahangeer S, Ritman EL, Russell SJ, Caplice NM. Intravascular cell delivery device for therapeutic VEGF-induced angiogenesis in chronic vascular occlusion. Biomaterials 2014; 35:9012-22. [PMID: 25096850 DOI: 10.1016/j.biomaterials.2014.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/10/2014] [Indexed: 10/24/2022]
Abstract
Site specific targeting remains elusive for gene and stem cell therapies in the cardiovascular field. One promising option involves use of devices that deliver larger and more sustained cell/gene payloads to specific disease sites using the versatility of percutaneous vascular access technology. Smooth muscle cells (SMCs) engineered to deliver high local concentrations of an angiogenic molecule (VEGF) were placed in an intravascular cell delivery device (ICDD) in a porcine model of chronic total occlusion (CTO) involving ameroid placement on the proximal left circumflex (LCx) artery. Implanted SMC were retained within the ICDD and were competent for VEGF production in vitro and in vivo. Following implantation, micro-CT analyses revealed that ICDD-VEGF significantly enhanced vasa vasora microvessel density with a concomitant increase in tissue VEGF protein levels and formation of endothelial cell colonies suggesting increased angiogenic potential. ICDD-VEGF markedly enhanced regional blood flow determined by microsphere and contrast CT analysis translating to a functional improvement in regional wall motion and global left ventricular (LV) systolic and diastolic function. Our data indicate robust, clinically relevant angiogenesis can be achieved in a human scale porcine chronic vascular occlusion model following ICDD-VEGF-based delivery of angiogenic cells. This may have implications for percutaneous delivery of numerous therapeutic factors promoting creation of microvascular bypass networks in chronic vaso-occlusive diseases.
Collapse
Affiliation(s)
- Arun H S Kumar
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Kenneth Martin
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Brendan Doyle
- Division of Cardiovascular Diseases, Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA
| | - Chien-Ling Huang
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Gopala-Krishnan M Pillai
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Mohammed T Ali
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Kimberly A Skelding
- Division of Cardiovascular Diseases, Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA
| | - Shaohua Wang
- Division of Cardiovascular Diseases, Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA
| | - Birgitta M Gleeson
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland
| | - Saleem Jahangeer
- Cork Cancer Research Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Erik L Ritman
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Stephen J Russell
- Division of Hematology, Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA
| | - Noel M Caplice
- Centre for Research in Vascular Biology (CRVB), Biosciences Institute, University College Cork, Cork, Ireland; Division of Cardiovascular Diseases, Molecular Medicine Program, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
70
|
Bruce AC, Kelly-Goss MR, Heuslein JL, Meisner JK, Price RJ, Peirce SM. Monocytes are recruited from venules during arteriogenesis in the murine spinotrapezius ligation model. Arterioscler Thromb Vasc Biol 2014; 34:2012-22. [PMID: 24969773 DOI: 10.1161/atvbaha.114.303399] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Chronic arterial occlusion results in arteriogenesis of collateral blood vessels. This process has been shown to be dependent on the recruitment of growth-promoting macrophages to remodeling collaterals. However, the potential role of venules in monocyte recruitment during microvascular arteriogenesis is not well demonstrated. First, we aim to document that arteriogenesis occurs in the mouse spinotrapezius ligation model. Then, we investigate the temporal and spatial distribution, as well as proliferation, of monocytes/macrophages recruited to collateral arterioles in response to elevated fluid shear stress. APPROACH AND RESULTS Laser speckle flowmetry confirmed a postligation increase in blood velocity within collateral arterioles but not within venules. After 72 hours post ligation, collateral arteriole diameters were increased, proliferating cells were identified in vessel walls of shear-activated collaterals, and perivascular CD206(+) macrophages demonstrated proliferation. A 5-ethynyl-2'-deoxyuridine assay identified proliferation. CD68(+)CD206(+) cells around collaterals were increased 96%, whereas CX3CR1((+/GFP)) cells were increased 126% in ligated versus sham groups after 72 hours. CX3CR1((+/GFP)) cells were predominately venule associated at 6 hours after ligation; and CX3CR1((+/GFP hi)) cells shifted from venule to arteriole associated between 6 and 72 hours after surgery exclusively in ligated muscle. We report accumulation and extravasation of adhered CX3CR1((+/GFP)) cells in and from venules, but not from arterioles, after ligation. CONCLUSIONS Our results demonstrate that arteriogenesis occurs in the murine spinotrapezius ligation model and implicate postcapillary venules as the site of tissue entry for circulating monocytes. Local proliferation of macrophages is also documented. These data open up questions about the role of arteriole-venule communication during monocyte recruitment.
Collapse
Affiliation(s)
- Anthony C Bruce
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville
| | - Molly R Kelly-Goss
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville
| | - Joshua L Heuslein
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville
| | - Joshua K Meisner
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville
| | - Richard J Price
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville
| | - Shayn M Peirce
- From the Department of Biomedical Engineering, University of Virginia, Charlottesville.
| |
Collapse
|
71
|
Bastiaansen AJNM, Karper JC, Wezel A, de Boer HC, Welten SMJ, de Jong RCM, Peters EAB, de Vries MR, van Oeveren-Rietdijk AM, van Zonneveld AJ, Hamming JF, Nossent AY, Quax PHA. TLR4 accessory molecule RP105 (CD180) regulates monocyte-driven arteriogenesis in a murine hind limb ischemia model. PLoS One 2014; 9:e99882. [PMID: 24945347 PMCID: PMC4063870 DOI: 10.1371/journal.pone.0099882] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/18/2014] [Indexed: 02/01/2023] Open
Abstract
AIMS We investigated the role of the TLR4-accessory molecule RP105 (CD180) in post-ischemic neovascularization, i.e. arteriogenesis and angiogenesis. TLR4-mediated activation of pro-inflammatory Ly6Chi monocytes is crucial for effective neovascularization. Immunohistochemical analyses revealed that RP105+ monocytes are present in the perivascular space of remodeling collateral arterioles. As RP105 inhibits TLR4 signaling, we hypothesized that RP105 deficiency would lead to an unrestrained TLR4-mediated inflammatory response and hence to enhanced blood flow recovery after ischemia. METHODS AND RESULTS RP105-/- and wild type (WT) mice were subjected to hind limb ischemia and blood flow recovery was followed by Laser Doppler Perfusion Imaging. Surprisingly, we found that blood flow recovery was severely impaired in RP105-/- mice. Immunohistochemistry showed that arteriogenesis was reduced in these mice compared to the WT. However, both in vivo and ex vivo analyses showed that circulatory pro-arteriogenic Ly6Chi monocytes were more readily activated in RP105-/- mice. FACS analyses showed that Ly6Chi monocytes became activated and migrated to the affected muscle tissues in WT mice following induction of hind limb ischemia. Although Ly6Chi monocytes were readily activated in RP105-/- mice, migration into the ischemic tissues was hampered and instead, Ly6Chi monocytes accumulated in their storage compartments, bone marrow and spleen, in RP105-/- mice. CONCLUSIONS RP105 deficiency results in an unrestrained inflammatory response and monocyte over-activation, most likely due to the lack of TLR4 regulation. Inappropriate, premature systemic activation of pro-inflammatory Ly6Chi monocytes results in reduced infiltration of Ly6Chi monocytes in ischemic tissues and in impaired blood flow recovery.
Collapse
Affiliation(s)
- Antonius J. N. M. Bastiaansen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacco C. Karper
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Anouk Wezel
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Hetty C. de Boer
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sabine M. J. Welten
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob C. M. de Jong
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Erna A. B. Peters
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Annemarie M. van Oeveren-Rietdijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaap F. Hamming
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - A. Yaël Nossent
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Paul H. A. Quax
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
72
|
van den Borne P, Rygiel TP, Hoogendoorn A, Westerlaken GHA, Boon L, Quax PHA, Pasterkamp G, Hoefer IE, Meyaard L. The CD200-CD200 receptor inhibitory axis controls arteriogenesis and local T lymphocyte influx. PLoS One 2014; 9:e98820. [PMID: 24897500 PMCID: PMC4045841 DOI: 10.1371/journal.pone.0098820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/07/2014] [Indexed: 12/05/2022] Open
Abstract
The role of the CD200 ligand-CD200 receptor (CD200-CD200R) inhibitory axis is highly important in controlling myeloid cell function. Since the activation of myeloid cells is crucial in arteriogenesis, we hypothesized that disruption of the CD200-CD200R axis promotes arteriogenesis in a murine hindlimb ischemia model. Female Cd200-/- and wildtype (C57Bl/6J) mice underwent unilateral femoral artery ligation. Perfusion recovery was monitored over 7 days using Laser-Doppler analysis and was increased in Cd200-/- mice at day 3 and 7 after femoral artery ligation, compared to wildtype. Histology was performed on hindlimb muscles at baseline, day 3 and 7 to assess vessel geometry and number and inflammatory cell influx. Vessel geometry in non-ischemic muscles was larger, and vessel numbers in ischemic muscles were increased in Cd200-/- mice compared to wildtype. Furthermore, T lymphocyte influx was increased in Cd200-/- compared to wildtype. CD200R agonist treatment was performed in male C57Bl/6J mice to validate the role of the CD200-CD200R axis in arteriogenesis. CD200R agonist treatment after unilateral femoral artery ligation resulted in a significant decrease in vessel geometry, perfusion recovery and T lymphocyte influx at day 7 compared to isotype treatment. In this study, we show a causal role for the CD200-CD200R inhibitory axis in arteriogenesis in a murine hindlimb ischemia model. Lack of CD200R signaling is accompanied by increased T lymphocyte recruitment to the collateral vasculature and results in enlargement of preexisting collateral arteries.
Collapse
Affiliation(s)
- Pleunie van den Borne
- Laboratory of Experimental Cardiology, University Medical Center, Utrecht, The Netherlands
| | - Tomasz P. Rygiel
- Laboratory for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Immunology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Ayla Hoogendoorn
- Laboratory of Experimental Cardiology, University Medical Center, Utrecht, The Netherlands
| | - Geertje H. A. Westerlaken
- Laboratory for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Paul H. A. Quax
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory of Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, University Medical Center, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Imo E. Hoefer
- Laboratory of Experimental Cardiology, University Medical Center, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Linde Meyaard
- Laboratory for Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
73
|
Herold J, Francke A, Weinert S, Schmeisser A, Hebel K, Schraven B, Roehl FW, Strasser RH, Braun-Dullaeus RC. Tetanus toxoid-pulsed monocyte vaccination for augmentation of collateral vessel growth. J Am Heart Assoc 2014; 3:e000611. [PMID: 24732919 PMCID: PMC4187481 DOI: 10.1161/jaha.113.000611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The pathogenesis of collateral growth (arteriogenesis) has been linked to both the innate and adaptive immune systems. While therapeutic approaches for the augmentation of arteriogenesis have focused on innate immunity, exploiting both innate and adaptive immune responses has not been examined. We hypothesized that tetanus toxoid (tt) immunization of mice followed by transplantation of monocytes (Mo) exposed ex vivo to tt augments arteriogenesis after ligation of the hind limb. Methods and Results Mo were generated from nonimmunized BALB/c mice, exposed ex vivo to tt for 24 hours and intravenously injected (ttMo, 2.5×106) into the tail veins of tt‐immunized syngeneic mice whose hind limbs had been ligated 24 hours prior to transplantation. Laser Doppler perfusion imaging was applied, and a perfusion index (PI) was calculated (ratio ligated/unligated). Twenty‐one days after ligation, the arteriogenesis of untreated BALB/c mice was limited (PI=0.49±0.09). Hind limb function was impaired in 80% of animals. Injection of non‐engineered Mo insignificantly increased the PI to 0.56±0.07. However, ttMo transplantation resulted in a strong increase of the PI to 0.82±0.08 (n=7; P<0.001), with no (0%) detectable functional impairment. ttMo injected into nonimmunized mice had no effect. The strong arteriogenic response of ttMo transplantation into immunized mice was prevented when mice had been depleted of T‐helper cells by CD4‐antibody pretreatment (PI=0.50±0.08; n=17; P<0.001), supporting the hypothesis that transplanted cells interact with recipient lymphocytes. Conclusions Transplantation of ttMo into pre‐immunized mice strongly promotes arteriogenesis. This therapeutic approach is feasible and highly attractive for the alleviation of morbidity associated with vascular occlusive disease.
Collapse
Affiliation(s)
- Joerg Herold
- Internal Medicine/Cardiology, Angiology and Pneumology, Magdeburg University, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Howangyin KY, Silvestre JS. Diabetes mellitus and ischemic diseases: molecular mechanisms of vascular repair dysfunction. Arterioscler Thromb Vasc Biol 2014; 34:1126-35. [PMID: 24675660 DOI: 10.1161/atvbaha.114.303090] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In patients with diabetes mellitus, the ability of ischemic tissue to synchronize the molecular and cellular events leading to restoration of tissue perfusion in response to the atherosclerotic occlusion of a patent artery is markedly impaired. As a consequence, adverse tissue remodeling and the extent of ischemic injury are intensified, leading to increased morbidity and mortality. Growing evidence from preclinical and clinical studies has implicated alterations in hypoxia-inducible factor 1 levels in the abrogation of proangiogenic pathways, including vascular endothelial growth factor A/phosphoinositide 3' kinase/AKT/endothelial nitric oxide synthase and in the activation of antiangiogenic signals characterized by accumulation of advanced glycation end products, reactive oxygen species overproduction, and endoplasmic reticulum stress. In addition, the diabetic milieu shows a switch toward proinflammatory antiregenerative pathways. Finally, the mobilization, subsequent recruitment, and the proangiogenic potential of the different subsets of angiogenesis-promoting bone marrow-derived cells are markedly impaired in the diabetic environment. In this review, we will give an overview of the current understanding on the signaling molecules contributing to the diabetes mellitus-induced impairment of postischemic revascularization mainly in the setting of myocardial infarction or critical limb ischemia.
Collapse
Affiliation(s)
- Kiave Yune Howangyin
- From the INSERM UMRS 970, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | | |
Collapse
|
75
|
Czepluch FS, Bernhardt M, Kuschicke H, Gogiraju R, Schroeter MR, Riggert J, Hasenfuss G, Schäfer K. In VitroandIn VivoEffects of Human Monocytes and their Subsets on New Vessel Formation. Microcirculation 2014; 21:148-58. [DOI: 10.1111/micc.12100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 10/07/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Frauke S. Czepluch
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Markus Bernhardt
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Hendrik Kuschicke
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Rajinikanth Gogiraju
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Marco R. Schroeter
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Joachim Riggert
- Department of Transfusion Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| | - Katrin Schäfer
- Department of Cardiology and Pulmonary Medicine; University Medical Center Göttingen; Göttingen Germany
| |
Collapse
|
76
|
Duelsner A, Gatzke N, Hillmeister P, Glaser J, Zietzer A, Nagorka S, Janke D, Pfitzner J, Stawowy P, Meyborg H, Urban D, Bondke Persson A, Buschmann IR. PPARγ activation inhibits cerebral arteriogenesis in the hypoperfused rat brain. Acta Physiol (Oxf) 2014; 210:354-68. [PMID: 24119262 DOI: 10.1111/apha.12179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 11/29/2022]
Abstract
AIMS PPARγ stimulation improves cardiovascular (CV) risk factors, but without improving overall clinical outcomes. PPARγ agonists interfere with endothelial cell (EC), monocyte and smooth muscle cell (SMC) activation, function and proliferation, physiological processes critical for arterial collateral growth (arteriogenesis). We therefore assessed the effect of PPARγ stimulation on cerebral adaptive and therapeutic collateral growth. METHODS In a rat model of adaptive cerebral arteriogenesis (3-VO), collateral growth and function were assessed (i) in controls, (ii) after PPARγ stimulation (pioglitazone 2.8 mg kg(-1); 10 mg kg(-1) compared with metformin 62.2 mg kg(-1) or sitagliptin 6.34 mg kg(-1)) for 21 days or (iii) after adding pioglitazone to G-CSF (40 μg kg(-1) every other day) to induce therapeutic arteriogenesis for 1 week. Pioglitazone effects on endothelial and SMC morphology and proliferation, monocyte activation and migration were studied. RESULTS PPARγ stimulation decreased cerebrovascular collateral growth and recovery of hemodynamic reserve capacity (CVRC controls: 12 ± 7%; pio low: -2 ± 9%; pio high: 1 ± 7%; metformin: 9 ± 13%; sitagliptin: 11 ± 12%), counteracted G-CSF-induced therapeutic arteriogenesis and interfered with EC activation, SMC proliferation, monocyte activation and migration. CONCLUSION Pharmacologic PPARγ stimulation inhibits pro-arteriogenic EC activation, monocyte function, SMC proliferation and thus adaptive as well as G-CSF-induced cerebral arteriogenesis. Further studies should evaluate whether this effect may underlie the CV risk associated with thiazolidinedione use in patients.
Collapse
Affiliation(s)
- A. Duelsner
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - N. Gatzke
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - P. Hillmeister
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - J. Glaser
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - A. Zietzer
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - S. Nagorka
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - D. Janke
- Julius Wolff Institute and Berlin-Brandenburg Center for Regenerative Therapies (CVK); Charité-Universitaetsmedizin Berlin; Berlin Germany
- Institute for Chemistry and Biochemistry; FU Berlin; Berlin Germany
| | - J. Pfitzner
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - P. Stawowy
- Department of Internal Medicine/Cardiology; German Heart Institute Berlin (DHZB); Berlin Germany
| | - H. Meyborg
- Department of Internal Medicine/Cardiology; German Heart Institute Berlin (DHZB); Berlin Germany
| | - D. Urban
- Department of Internal Medicine/Cardiology; German Heart Institute Berlin (DHZB); Berlin Germany
| | - A. Bondke Persson
- Institute of Vegetative Physiology; Charité - Universitaetsmedizin Berlin; Berlin Germany
| | - I. R. Buschmann
- Center for Cardiovascular Research (CCR); Richard-Thoma-Laboratories for Arteriogenesis; Charité - Universitaetsmedizin Berlin; Berlin Germany
| |
Collapse
|
77
|
van den Borne P, Haverslag RT, Brandt MM, Cheng C, Duckers HJ, Quax PHA, Hoefer IE, Pasterkamp G, de Kleijn DPV. Absence of chemokine (C-x-C motif) ligand 10 diminishes perfusion recovery after local arterial occlusion in mice. Arterioscler Thromb Vasc Biol 2014; 34:594-602. [PMID: 24407030 DOI: 10.1161/atvbaha.113.303050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In arteriogenesis, pre-existing anastomoses undergo enlargement to restore blood flow in ischemic tissues. Chemokine (C-X-C motif) ligand 10 (CXCL10) is secreted after Toll-like receptor activation. Toll-like receptors are involved in arteriogenesis; however, the role of CXCL10 is still unclear. In this study, we investigated the role for CXCL10 in a murine hindlimb ischemia model. APPROACH AND RESULTS Unilateral femoral artery ligation was performed in wild-type (WT) and CXCL10(-/-) knockout (KO) mice and perfusion recovery was measured using laser-Doppler perfusion analysis. Perfusion recovery was significantly lower in KO mice compared with WT at days 4 and 7 after surgery (KO versus WT: 28±5% versus 81±13% at day 4; P=0.003 and 57±12% versus 107±8% at day 7; P=0.003). Vessel measurements of α-smooth muscle actin-positive vessels revealed increasing numbers in time after surgery, which was significantly higher in WT when compared with that in KO. Furthermore, α-smooth muscle actin-positive vessels were significantly larger in WT when compared with those in KO at day 7 (wall thickness, P<0.001; lumen area, P=0.003). Local inflammation was assessed in hindlimb muscles, but this did not differ between WT and KO. Chimerization experiments analyzing perfusion recovery and histology revealed an equal contribution for bone marrow-derived and circulating CXCL10. Migration assays showed a stimulating role for both intrinsic and extrinsic CXCL10 in vascular smooth muscle cell migration. CONCLUSIONS CXCL10 plays a causal role in arteriogenesis. Bone marrow-derived CXCL10 and tissue-derived CXCL10 play a critical role in accelerating perfusion recovery after arterial occlusion in mice probably by promoting vascular smooth muscle cell recruitment and maturation of pre-existing anastomoses.
Collapse
Affiliation(s)
- Pleunie van den Borne
- From the Laboratory of Experimental Cardiology (P.v.d.B., R.T.H., I.E.H., G.P., D.P.V.d.K.), Department of Nephrology and Hypertension (C.C.), and Department of Cardiology (H.J.D.), University Medical Center Utrecht, Utrecht, The Netherlands; Molecular Cardiology Laboratory, Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands (M.M.B., C.C.); Department of Surgery (P.H.A.Q.) and Einthoven Laboratory of Experimental Vascular Medicine (P.H.A.Q.), Leiden University Medical Center, Leiden, The Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (I.E.H., G.P., D.P.V.d.K.); and Cardiovascular Research Institute and Surgery, National University Hospital, Singapore, Singapore (D.P.V.d.K.)
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
79
|
Liu J, Wang Y, Akamatsu Y, Lee CC, Stetler RA, Lawton MT, Yang GY. Vascular remodeling after ischemic stroke: mechanisms and therapeutic potentials. Prog Neurobiol 2013; 115:138-56. [PMID: 24291532 DOI: 10.1016/j.pneurobio.2013.11.004] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 11/14/2013] [Accepted: 11/16/2013] [Indexed: 12/18/2022]
Abstract
The brain vasculature has been increasingly recognized as a key player that directs brain development, regulates homeostasis, and contributes to pathological processes. Following ischemic stroke, the reduction of blood flow elicits a cascade of changes and leads to vascular remodeling. However, the temporal profile of vascular changes after stroke is not well understood. Growing evidence suggests that the early phase of cerebral blood volume (CBV) increase is likely due to the improvement in collateral flow, also known as arteriogenesis, whereas the late phase of CBV increase is attributed to the surge of angiogenesis. Arteriogenesis is triggered by shear fluid stress followed by activation of endothelium and inflammatory processes, while angiogenesis induces a number of pro-angiogenic factors and circulating endothelial progenitor cells (EPCs). The status of collaterals in acute stroke has been shown to have several prognostic implications, while the causal relationship between angiogenesis and improved functional recovery has yet to be established in patients. A number of interventions aimed at enhancing cerebral blood flow including increasing collateral recruitment are under clinical investigation. Transplantation of EPCs to improve angiogenesis is also underway. Knowledge in the underlying physiological mechanisms for improved arteriogenesis and angiogenesis shall lead to more effective therapies for ischemic stroke.
Collapse
Affiliation(s)
- Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA.
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai 200030, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yosuke Akamatsu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurological Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Chih Cheng Lee
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Michael T Lawton
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai 200030, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| |
Collapse
|
80
|
Abstract
Vasculogenesis and angiogenesis are the major forms of blood vessel formation. Angiogenesis is the process where new vessels grow from pre-existing blood vessels, and is very important in the functional recovery of pathological conditions, such as wound healing and ischemic heart diseases. The development of better animal model and imaging technologies in past decades has greatly enriched our understanding on vasculogenesis and angiogenesis processes. Hypoxia turned out to be an important driving force for angiogenesis in various ischemic conditions. It stimulates expression of many growth factors like vascular endothelial growth factor, platelet-derived growth factor, insulin-like growth factor, and fibroblast growth factor, which play critical role in induction of angiogenesis. Other cellular components like monocytes, T cells, neutrophils, and platelets also play significant role in induction and regulation of angiogenesis. Various stem/progenitor cells also being recruited to the ischemic sites play crucial role in the angiogenesis process. Pre-clinical studies showed that stem/progenitor cells with/without combination of growth factors induce neovascularization in the ischemic tissues in various animal models. In this review, we will discuss about the fundamental factors that regulate the angiogenesis process and the use of stem cells as therapeutic regime for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Jingwei Lu
- Cardiovascular Stem Cell Research Laboratory, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, 460W, 12th Avenue, BRT 382, Columbus, OH, 43210, USA
| | | | | |
Collapse
|
81
|
Robbins CS, Hilgendorf I, Weber GF, Theurl I, Iwamoto Y, Figueiredo JL, Gorbatov R, Sukhova GK, Gerhardt LMS, Smyth D, Zavitz CCJ, Shikatani EA, Parsons M, van Rooijen N, Lin HY, Husain M, Libby P, Nahrendorf M, Weissleder R, Swirski FK. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med 2013; 19:1166-72. [PMID: 23933982 PMCID: PMC3769444 DOI: 10.1038/nm.3258] [Citation(s) in RCA: 775] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/29/2013] [Indexed: 12/22/2022]
Abstract
During the inflammatory response that drives atherogenesis, macrophages accumulate progressively in the expanding arterial wall1,2. The observation that circulating monocytes give rise to lesional macrophages3–9 has reinforced the concept that monocyte infiltration dictates macrophage build-up. Recent work indicates, however, that macrophages do not depend on monocytes in some inflammatory contexts10. We therefore revisited the mechanism of macrophage accumulation in atherosclerosis. We show that murine atherosclerotic lesions experience a surprisingly rapid, 4-week, cell turnover. Replenishment of macrophages in these experimental atheromata depends predominantly on local macrophage proliferation rather than monocyte influx. The microenvironment orchestrates macrophage proliferation via the involvement of scavenger receptor (SR)-A. Our study reveals macrophage proliferation as a key event in atherosclerosis and identifies macrophage self-renewal as a therapeutic target for cardiovascular disease.
Collapse
Affiliation(s)
- Clinton S Robbins
- 1] Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada. [3] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. [4] Department of Immunology, University of Toronto, Toronto, Ontario, Canada. [5]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Local delivery of polarized macrophages improves reperfusion recovery in a mouse hind limb ischemia model. PLoS One 2013; 8:e68811. [PMID: 23894348 PMCID: PMC3722193 DOI: 10.1371/journal.pone.0068811] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/31/2013] [Indexed: 12/31/2022] Open
Abstract
AIMS Enhancement of collateral development in coronary or peripheral artery disease is a therapeutic target, but it has proven difficult to achieve. Macrophages are key players in collateral remodeling, yet the effect of different macrophage subsets on arteriogenesis has not been investigated. METHODS AND RESULTS Murine macrophages were cultured from bone marrow and polarized into M1 (IFNγ), M2a (IL-4) or M2c (IL-10) subsets. C57BL/6 mice underwent femoral artery ligation followed by intramuscular injection of macrophage subsets. Using eGFP expressing macrophages, cells could be detected at least 6 days after ligation and were located in the perivascular space of collateral vessels. After 14 days, perfusion ratio was increased in animals treated with M1 as well as M2a and M2c macrophages compared to control. Depletion of circulating monocytes by clodronate liposome injections did not hamper reperfusion recovery, however, treatment with exogenous polarized macrophages improved perfusion ratio after 14 days again. We used IL10R(fl/fl)/LysMCre(+) mice to study the effect of inhibition of endogenous polarization towards specifically M2c macrophages on arteriogenesis. Deletion of the IL10-receptor (IL10R) in the myeloid lineage did not affect reperfusion recovery, yet the pro-arteriogenic effect of exogenously injected M2c macrophages was still present. CONCLUSIONS Local injection of polarized macrophages promotes reperfusion recovery after femoral artery ligation and is not influenced by depletion of circulatory monocytes. Preventing endogenous M2c polarization did not affect reperfusion recovery suggesting that M2c's are not required for collateralization, but are sufficient to induce collateral formation upon exogenous administration. This is the first study using local injection of macrophage subsets showing the pro-arteriogenic effect of polarized macrophages.
Collapse
|
83
|
Ouma GO, Rodriguez E, Muthumani K, Weiner DB, Wilensky RL, Mohler ER. In vivo electroporation of constitutively expressed HIF-1α plasmid DNA improves neovascularization in a mouse model of limb ischemia. J Vasc Surg 2013; 59:786-93. [PMID: 23850058 DOI: 10.1016/j.jvs.2013.04.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/09/2013] [Accepted: 04/17/2013] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor that stimulates angiogenesis during tissue ischemia. In vivo electroporation (EP) enhances tissue DNA transfection. We hypothesized that in vivo EP of plasmid DNA encoding a constitutively expressed HIF-1α gene enhances neovascularization compared with intramuscular (IM) injection alone. METHODS Left femoral artery ligation was performed in mice assigned to three groups: (1) HIF-EP (n = 13); (2) HIF-IM (n = 14); and (3) empty plasmid (pVAX)-EP (n = 12). A single dose of HIF-1α or pVAX DNA (20 μL of 5 μg/μL each) was injected into the ischemic adductor muscle followed by EP (groups one and three). Mice in group two received IM injection of HIF-1α plasmid DNA alone. From preligation to days 0, 3, 7, 14, and 21 postligation, limb perfusion recovery quantified by laser Doppler perfusion imager, limb function, and limb necrosis were measured. On day 21, the surviving mice (4-5 per group) were sacrificed and adductor muscle tissues stained for necrosis using hematoxylin and eosin, capillary density (anti-CD31 antibodies), and collateral vessels via anti-α-smooth muscle actin antibodies. RESULTS In vivo EP of HIF-1α DNA significantly improved limb perfusion (HIF-EP: 1.03 ± 0.15 vs HIF-IM: 0.78 ± 0.064; P < .05, vs pVAX-EP: 0.41 ± 0.019; P < .001), limb functional recovery (HIF-EP: 3.5 ± 0.58 vs HIF-IM, 2.4 ± 1.14; P < .05, vs pVAX-EP: 2.4 ± 1.14; P < .001), and limb autoamputation on day 21 (HIF-EP: 77% ± 12% vs HIF-IM: 43% ± 14%; P < .05 vs pVAX-EP: 17% ± 11%; P < .01). Adductor muscle tissue necrosis decreased (HIF-EP: 20.7% ± 1.75% vs HIF-IM: 44% ± 3.73; P < .001, vs pVAX-EP: 60.05% ± 2.17%; P < .0001), capillary density increased (HIF-EP: 96.83 ± 5.72 vessels/high-powered field [hpf] vs HIF-IM: 62.87 ± 2.0 vessels/hpf; P < .001, vs pVAX-EP: 39.37 ± 2.76 vessels/hpf; P < .0001), collateral vessel formation increased (HI-EP: 76.33 ± 1.94 vessels/hpf vs HIF-IM: 37.5 ± 1.56 vessels/hpf; P < .0001, vs pVAX-EP: 18.5 ± 1.34 vessels/hpf; P < .00001), and the vessels were larger (HIF-EP: 15,521.67 ± 1298.16 μm(2) vs HIF-IM: 7788.87 ± 392.04 μm(2); P < .001 vs pVAX-EP: 4640.25 ± 614.01 μm(2); P < .0001). CONCLUSIONS In vivo EP-mediated delivery of HIF-1α plasmid DNA improves neovascularization in a mouse model of limb ischemia and is a potentially suitable nonviral, noninvasive intervention to facilitate therapeutic angiogenesis in critical limb ischemia.
Collapse
Affiliation(s)
- Geoffrey O Ouma
- Department of Medicine, Cardiovascular Division, Vascular Medicine Section, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa.
| | - Eduardo Rodriguez
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa
| | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa
| | - Robert L Wilensky
- Cardiovascular Division, Hospitial of the University of Pennsylvania and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa
| | - Emile R Mohler
- Department of Medicine, Cardiovascular Division, Vascular Medicine Section, Perelman School of Medicine at the University of Pennsylvania and University of Pennsylvania Health System, Philadelphia, Pa
| |
Collapse
|
84
|
Cubbon RM, Mercer BN, Sengupta A, Kearney MT. Importance of insulin resistance to vascular repair and regeneration. Free Radic Biol Med 2013; 60:246-63. [PMID: 23466555 DOI: 10.1016/j.freeradbiomed.2013.02.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/22/2013] [Accepted: 02/23/2013] [Indexed: 01/14/2023]
Abstract
Metabolic insulin resistance is apparent across a spectrum of clinical disorders, including obesity and diabetes, and is characterized by an adverse clustering of cardiovascular risk factors related to abnormal cellular responses to insulin. These disorders are becoming increasingly prevalent and represent a major global public health concern because of their association with significant increases in atherosclerosis-related mortality. Endogenous repair mechanisms are thought to retard the development of vascular disease, and a growing evidence base supports the adverse impact of the insulin-resistant phenotype upon indices of vascular repair. Beyond the impact of systemic metabolic changes, emerging data from murine studies also provide support for abnormal insulin signaling at the level of vascular cells in retarding vascular repair. Interrelated pathophysiological factors, including reduced nitric oxide bioavailability, oxidative stress, altered growth factor activity, and abnormal intracellular signaling, are likely to act in conjunction to impede vascular repair while also driving vascular damage. Understanding of these processes is shaping novel therapeutic paradigms that aim to promote vascular repair and regeneration, either by recruiting endogenous mechanisms or by the administration of cell-based therapies.
Collapse
Affiliation(s)
- Richard M Cubbon
- Multidisciplinary Cardiovascular Research Centre, LIGHT Laboratories, The University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | |
Collapse
|
85
|
Cunha FFD, Martins L, Martin PKM, Stilhano RS, Paredes Gamero EJ, Han SW. Comparison of treatments of peripheral arterial disease with mesenchymal stromal cells and mesenchymal stromal cells modified with granulocyte and macrophage colony-stimulating factor. Cytotherapy 2013; 15:820-9. [DOI: 10.1016/j.jcyt.2013.02.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 02/20/2013] [Accepted: 02/25/2013] [Indexed: 01/26/2023]
|
86
|
Bastiaansen AJNM, Ewing MM, de Boer HC, van der Pouw Kraan TC, de Vries MR, Peters EAB, Welten SMJ, Arens R, Moore SM, Faber JE, Jukema JW, Hamming JF, Nossent AY, Quax PHA. Lysine acetyltransferase PCAF is a key regulator of arteriogenesis. Arterioscler Thromb Vasc Biol 2013; 33:1902-10. [PMID: 23788761 DOI: 10.1161/atvbaha.113.301579] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Therapeutic arteriogenesis, that is, expansive remodeling of preexisting collaterals, using single-action factor therapies has not been as successful as anticipated. Modulation of factors that act as a master switch for relevant gene programs may prove more effective. Transcriptional coactivator p300-CBP-associated factor (PCAF) has histone acetylating activity and promotes transcription of multiple inflammatory genes. Because arteriogenesis is an inflammation-driven process, we hypothesized that PCAF acts as multifactorial regulator of arteriogenesis. APPROACH AND RESULTS After induction of hindlimb ischemia, blood flow recovery was impaired in both PCAF(-/-) mice and healthy wild-type mice treated with the pharmacological PCAF inhibitor Garcinol, demonstrating an important role for PCAF in arteriogenesis. PCAF deficiency reduced the in vitro inflammatory response in leukocytes and vascular cells involved in arteriogenesis. In vivo gene expression profiling revealed that PCAF deficiency results in differential expression of 3505 genes during arteriogenesis and, more specifically, in impaired induction of multiple proinflammatory genes. Additionally, recruitment from the bone marrow of inflammatory cells, in particular proinflammatory Ly6C(hi) monocytes, was severely impaired in PCAF(-/-) mice. CONCLUSIONS These findings indicate that PCAF acts as master switch in the inflammatory processes required for effective arteriogenesis.
Collapse
Affiliation(s)
- Antonius J N M Bastiaansen
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Hamm A, Veschini L, Takeda Y, Costa S, Delamarre E, Squadrito ML, Henze AT, Wenes M, Serneels J, Pucci F, Roncal C, Anisimov A, Alitalo K, De Palma M, Mazzone M. PHD2 regulates arteriogenic macrophages through TIE2 signalling. EMBO Mol Med 2013; 5:843-57. [PMID: 23616286 PMCID: PMC3779447 DOI: 10.1002/emmm.201302695] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 03/09/2013] [Accepted: 03/12/2013] [Indexed: 01/26/2023] Open
Abstract
Occlusion of the main arterial route redirects blood flow to the collateral circulation. We previously reported that macrophages genetically modified to express low levels of prolyl hydroxylase domain protein 2 (PHD2) display an arteriogenic phenotype, which promotes the formation of collateral vessels and protects the skeletal muscle from ischaemic necrosis. However, the molecular mechanisms underlying this process are unknown. Here, we demonstrate that femoral artery occlusion induces a switch in macrophage phenotype through angiopoietin-1 (ANG1)-mediated Phd2 repression. ANG blockade by a soluble trap prevented the downregulation of Phd2 expression in macrophages and their phenotypic switch, thus inhibiting collateral growth. ANG1-dependent Phd2 repression initiated a feed-forward loop mediated by the induction of the ANG receptor TIE2 in macrophages. Gene silencing and cell depletion strategies demonstrate that TIE2 induction in macrophages is required to promote their proarteriogenic functions, enabling collateral vessel formation following arterial obstruction. These results indicate an indispensable role for TIE2 in sustaining in situ programming of macrophages to a proarteriogenic, M2-like phenotype, suggesting possible new venues for the treatment of ischaemic disorders.
Collapse
Affiliation(s)
- Alexander Hamm
- Laboratory of Molecular Oncology and Angiogenesis, Vesalius Research Center, VIB, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
|
89
|
Abstract
SIGNIFICANCE Proangiogenic therapy appeared a promising strategy for the treatment of patients with acute myocardial infarction (MI), as de novo formation of microvessels, has the potential to salvage ischemic myocardium at early stages after MI, and is also essential to prevent the transition to heart failure through the control of cardiomyocyte hypertrophy and contractility. RECENT ADVANCES Exciting preclinical studies evaluating proangiogenic therapies for MI have prompted the initiation of numerous clinical trials based on protein or gene transfer delivery of growth factors and administration of stem/progenitor cells, mainly from bone marrow origin. Nonetheless, these clinical trials showed mixed results in patients with acute MI. CRITICAL ISSUES Even though methodological caveats, such as way of delivery for angiogenic growth factors (e.g., protein vs. gene transfer) and stem/progenitor cells or isolation/culture procedure for regenerative cells might partially explain the failure of such trials, it appears that delivery of a single growth factor or cell type does not support angiogenesis sufficiently to promote cardiac repair. FUTURE DIRECTIONS Optimization of proangiogenic therapies might include stimulation of both angiogenesis and vessel maturation and/or the use of additional sources of stem/progenitor cells, such as cardiac progenitor cells. Experimental unraveling of the mechanisms of angiogenesis, vessel maturation, and endothelial cell/cardiomyocyte cross talk in the ischemic heart, analysis of emerging pathways, as well as a better understanding of how cardiovascular risk factors impact endogenous and therapeutically stimulated angiogenesis, would undoubtedly pave the way for the development of novel and hopefully efficient angiogenesis targeting therapeutics for the treatment of acute MI.
Collapse
Affiliation(s)
- Clement Cochain
- Paris Cardiovascular Research Center, INSERM UMR-S 970, Paris Descartes University, Paris, France
| | | | | |
Collapse
|
90
|
|
91
|
Abstract
BACKGROUND The role of bone marrow-derived cells in stimulating angiogenesis, vascular repair or remodelling has been well established, but the nature of the circulating angiogenic cells is still controversial. DESIGN The existing literature on different cell types that contribute to angiogenesis in multiple pathologies, most notably ischaemic and tumour angiogenesis, is reviewed, with a focus on subtypes of angiogenic mononuclear cells and their local recruitment and activation. RESULTS A large number of different cells of myeloid origin support angiogenesis without incorporating permanently into the newly formed vessel, which distinguishes these circulating angiogenic cells (CAC) from endothelial progenitor cells (EPC). Although CAC frequently express individual endothelial markers, they all share multiple characteristics of monocytes and only express a limited set of discriminative surface markers in the circulation. When cultured ex vivo, or surrounding the angiogenic vessel in vivo, however, many of them acquire similar additional markers, making their discrimination in situ difficult. CONCLUSION Different subsets of monocytes show angiogenic properties, but the distinct microenvironment, in vitro or in vivo, is needed for the development of their pro-angiogenic function.
Collapse
Affiliation(s)
- Julie Favre
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | |
Collapse
|
92
|
Abstract
Peripheral arterial disease (PAD) is a common vascular disease that reduces blood flow capacity to the legs of patients. PAD leads to exercise intolerance that can progress in severity to greatly limit mobility, and in advanced cases leads to frank ischemia with pain at rest. It is estimated that 12 to 15 million people in the United States are diagnosed with PAD, with a much larger population that is undiagnosed. The presence of PAD predicts a 50% to 1500% increase in morbidity and mortality, depending on severity. Treatment of patients with PAD is limited to modification of cardiovascular disease risk factors, pharmacological intervention, surgery, and exercise therapy. Extended exercise programs that involve walking approximately five times per week, at a significant intensity that requires frequent rest periods, are most significant. Preclinical studies and virtually all clinical trials demonstrate the benefits of exercise therapy, including improved walking tolerance, modified inflammatory/hemostatic markers, enhanced vasoresponsiveness, adaptations within the limb (angiogenesis, arteriogenesis, and mitochondrial synthesis) that enhance oxygen delivery and metabolic responses, potentially delayed progression of the disease, enhanced quality of life indices, and extended longevity. A synthesis is provided as to how these adaptations can develop in the context of our current state of knowledge and events known to be orchestrated by exercise. The benefits are so compelling that exercise prescription should be an essential option presented to patients with PAD in the absence of contraindications. Obviously, selecting for a lifestyle pattern that includes enhanced physical activity prior to the advance of PAD limitations is the most desirable and beneficial.
Collapse
Affiliation(s)
- Tara L Haas
- Angiogenesis Research Group, Muscle Health Research Centre, Faculty of Health, York University, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
93
|
Santini MP, Rosenthal N. Myocardial regenerative properties of macrophage populations and stem cells. J Cardiovasc Transl Res 2012; 5:700-12. [PMID: 22684511 PMCID: PMC3447141 DOI: 10.1007/s12265-012-9383-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/24/2012] [Indexed: 01/02/2023]
Abstract
The capacity to regenerate damaged tissue and appendages is lost to some extent in higher vertebrates such as mammals, which form a scar tissue at the expenses of tissue reconstitution and functionality. Whereas this process can protect from further damage and elicit fast healing, it can lead to functional deterioration in organs such as the heart. Based on the analyses performed in the last years, stem cell therapies may not be sufficient to induce cardiac regeneration and additional approaches are required to overcome scar formation. Among these, the immune cells and their humoral response have become a key parameter in regenerative processes. In this review, we will describe the recent findings on the possible therapeutical use of progenitor and immune cells to rescue a damaged heart.
Collapse
|
94
|
Abstract
Arteriosclerotic vascular disease is the most common cause of death and a major cause of disability in the developed world. Adverse outcomes of arteriosclerotic vascular disease are related to consequences of tissue ischemia and necrosis affecting the heart, brain, limbs, and other organs. Collateral artery growth or arteriogenesis occurs naturally and can help restore perfusion to ischemic tissues. Understanding the mechanisms of collateral artery growth may provide therapeutic options for patients with ischemic vascular disease. In this review, we examine the evidence for a role of monocytes and macrophages in collateral arteriogenesis.
Collapse
Affiliation(s)
- Erik Fung
- Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical CenterLebanon, NH, USA
| | - Armin Helisch
- Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical CenterLebanon, NH, USA
| |
Collapse
|
95
|
Böring YC, Flögel U, Jacoby C, Heil M, Schaper W, Schrader J. Lack of ecto-5'-nucleotidase (CD73) promotes arteriogenesis. Cardiovasc Res 2012; 97:88-96. [PMID: 22977005 DOI: 10.1093/cvr/cvs286] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Adenosine can stimulate angiogenesis, but its role in the distinct process of arteriogenesis is unknown. We have previously reported that mice lacking ecto-5'-nucleotidase (CD73-/-) show enhanced monocyte adhesion to the endothelium after ischaemia, which is considered to be an important trigger for arteriogenesis. METHODS AND RESULTS Hindlimb ischaemia was induced in wild-type (WT) and CD73-/- mice to study the role of extracellularly formed adenosine in arteriogenesis. Magnetic resonance angiography (MRA) was performed for serial visualization of newly developed vessels at a spatial resolution of 1 nL, and high-energy phosphates (HEP) were quantified by (31)P MR spectroscopy (MRS). MRA of CD73-/- mice revealed substantially enhanced collateral artery conductance at day 7 [CD73-/-: 0.73 ± 0.11 a.u. (arbitrary units); WT: 0.44 ± 0.13 a.u.; P < 0.01, n = 6], and MRS of the affected hindlimb showed a faster restoration of HEP in correlation with enhanced functional recovery in the mutant. Additionally, histology showed no differences in capillary density between the groups but showed an increased monocyte infiltration in hindlimbs of CD73-/- mice. CONCLUSION Serial assessment of dynamic changes of vessel growth and metabolism in the process of arteriogenesis demonstrate that the lack of CD73-derived adenosine importantly promotes arteriogenesis but does not alter angiogenesis in our model of hindlimb ischaemia.
Collapse
Affiliation(s)
- Yang Chul Böring
- Department of Molecular Cardiology, Heinrich Heine University of Düsseldorf, Moorenstr. 5, Düsseldorf 40225, Germany
| | | | | | | | | | | |
Collapse
|
96
|
Pardali E, Waltenberger J. Monocyte function and trafficking in cardiovascular disease. Thromb Haemost 2012; 108:804-11. [PMID: 22918193 DOI: 10.1160/th12-04-0276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/24/2012] [Indexed: 01/13/2023]
Abstract
Monocytes are key effectors of the immune homeostasis and play a crucial role in (vascular) injury repair. Despite their role in immune defense and tissue repair mechanisms, monocytes are also involved in several pathological conditions such as autoimmune and cardiovascular diseases as well as cancer. This suggests that monocytes can be used as diagnostic and as therapeutic targets. A better understanding and characterisation of monocytes and their function in both physiological and pathological situations is thus of great interest. This review focuses on recent advances on the role of monocytes in cardiovascular diseases and describes the value of monocytes as either disease marker or therapeutic target for (cardio)vascular diseases.
Collapse
Affiliation(s)
- Evangelia Pardali
- Evangelia Pardali or Johannes Waltenberger, Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer-Campus 1, Building A1, 48149 Münster, Germany.
| | | |
Collapse
|
97
|
Regulation of collateral blood vessel development by the innate and adaptive immune system. Trends Mol Med 2012; 18:494-501. [PMID: 22818027 DOI: 10.1016/j.molmed.2012.06.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/11/2012] [Accepted: 06/15/2012] [Indexed: 12/21/2022]
|
98
|
Landázuri N, Joseph G, Guldberg RE, Taylor WR. Growth and regression of vasculature in healthy and diabetic mice after hindlimb ischemia. Am J Physiol Regul Integr Comp Physiol 2012; 303:R48-56. [PMID: 22573106 DOI: 10.1152/ajpregu.00002.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The formation of vascular networks during embryogenesis and early stages of development encompasses complex and tightly regulated growth of blood vessels, followed by maturation of some vessels, and spatially controlled disconnection and pruning of others. The adult vasculature, while more quiescent, is also capable of adapting to changing physiological conditions by remodeling blood vessels. Numerous studies have focused on understanding key factors that drive vessel growth in the adult in response to ischemic injury. However, little is known about the extent of vessel rarefaction and its potential contribution to the final outcome of vascular recovery. We addressed this topic by characterizing the endogenous phases of vascular repair in a mouse model of hindlimb ischemia. We showed that this process is biphasic. It encompasses an initial rapid phase of vessel growth, followed by a later phase of vessel rarefaction. In healthy mice, this process resulted in partial recovery of perfusion and completely restored the ability of mice to run voluntarily. Given that the ability to revascularize can be compromised by a cardiovascular risk factor such as diabetes, we also examined vascular repair in diabetic mice. We found that paradoxically both the initial growth and subsequent regression of collateral vessels were more pronounced in the setting of diabetes and resulted in impaired recovery of perfusion and impaired functional status. In conclusion, our findings demonstrate that the formation of functional collateral vessels in the hindlimb requires vessel growth and subsequent vessel rarefaction. In the setting of diabetes, the physiological defect was not in the initial formation of vessels but rather in the inability to sustain newly formed vessels.
Collapse
Affiliation(s)
- Natalia Landázuri
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | |
Collapse
|
99
|
Skuli N, Majmundar AJ, Krock BL, Mesquita RC, Mathew LK, Quinn ZL, Runge A, Liu L, Kim MN, Liang J, Schenkel S, Yodh AG, Keith B, Simon MC. Endothelial HIF-2α regulates murine pathological angiogenesis and revascularization processes. J Clin Invest 2012; 122:1427-43. [PMID: 22426208 PMCID: PMC3314446 DOI: 10.1172/jci57322] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/03/2012] [Indexed: 12/12/2022] Open
Abstract
Localized tissue hypoxia is a consequence of vascular compromise or rapid cellular proliferation and is a potent inducer of compensatory angiogenesis. The oxygen-responsive transcriptional regulator hypoxia-inducible factor 2α (HIF-2α) is highly expressed in vascular ECs and, along with HIF-1α, activates expression of target genes whose products modulate vascular functions and angiogenesis. However, the mechanisms by which HIF-2α regulates EC function and tissue perfusion under physiological and pathological conditions are poorly understood. Using mice in which Hif2a was specifically deleted in ECs, we demonstrate here that HIF-2α expression is required for angiogenic responses during hindlimb ischemia and for the growth of autochthonous skin tumors. EC-specific Hif2a deletion resulted in increased vessel formation in both models; however, these vessels failed to undergo proper arteriogenesis, resulting in poor perfusion. Analysis of cultured HIF-2α-deficient ECs revealed cell-autonomous increases in migration, invasion, and morphogenetic activity, which correlated with HIF-2α-dependent expression of specific angiogenic factors, including delta-like ligand 4 (Dll4), a Notch ligand, and angiopoietin 2. By stimulating Dll4 signaling in cultured ECs or restoring Dll4 expression in ischemic muscle tissue, we rescued most of the HIF-2α-dependent EC phenotypes in vitro and in vivo, emphasizing the critical role of Dll4/Notch signaling as a downstream target of HIF-2α in ECs. These results indicate that HIF-1α and HIF-2α fulfill complementary, but largely nonoverlapping, essential functions in pathophysiological angiogenesis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Angiopoietin-2/genetics
- Angiopoietin-2/physiology
- Animals
- Basic Helix-Loop-Helix Transcription Factors/deficiency
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/physiology
- Calcium-Binding Proteins
- Cell Hypoxia
- Cell Movement
- Cells, Cultured/cytology
- Collateral Circulation/physiology
- Endothelial Cells/metabolism
- Hindlimb/blood supply
- Hypoxia-Inducible Factor 1, alpha Subunit/deficiency
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/physiology
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/physiology
- Ischemia/physiopathology
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/physiology
- Receptors, Notch/physiology
- Recombinant Fusion Proteins/physiology
- Recovery of Function
- Skin Neoplasms/blood supply
- Skin Neoplasms/chemically induced
- Wound Healing/physiology
Collapse
Affiliation(s)
- Nicolas Skuli
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amar J. Majmundar
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bryan L. Krock
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rickson C. Mesquita
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lijoy K. Mathew
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zachary L. Quinn
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anja Runge
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Liping Liu
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meeri N. Kim
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jiaming Liang
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Steven Schenkel
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arjun G. Yodh
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian Keith
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M. Celeste Simon
- Howard Hughes Medical Institute,
Abramson Family Cancer Research Institute,
School of Medicine,
Department of Physics and Astronomy,
Department of Cancer Biology, and
Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
100
|
Fefer P, Robert N, Qiang B, Liu G, Munce N, Anderson K, Osherov A, Ladouceur-Wodzak M, Qi X, Dick A, Weisbrod M, Samuel M, Butany J, Wright G, Strauss B. Characterisation of a novel porcine coronary artery CTO model. EUROINTERVENTION 2012; 7:1444-52. [DOI: 10.4244/eijv7i12a225] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|