Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicolle Kränkel is active.

Publication


Featured researches published by Nicolle Kränkel.


Circulation | 2005

Impact of Regular Physical Activity on the NAD(P)H Oxidase and Angiotensin Receptor System in Patients With Coronary Artery Disease

Volker Adams; Axel Linke; Nicolle Kränkel; Sandra Erbs; Stephan Gielen; Sven Möbius-Winkler; Jan Gummert; Friedrich W. Mohr; Gerhard Schuler; Rainer Hambrecht

Background—In patients with stable coronary artery disease, physical exercise training (ET) improves endothelial dysfunction. A potential mechanism mediating the enhanced vasomotor function is a reduced breakdown of endothelium-derived nitric oxide by reactive oxygen species (ROS). The aim of the present study was to analyze the impact of ET on sources of ROS generation in the left internal mammary artery of patients with symptomatic coronary artery disease. Methods and Results—In left internal mammary artery rings sampled during bypass surgery from 45 patients randomized to either a training (n=22) or an inactive control (n=23) group, the mRNA expression of NAD(P)H oxidase subunits, NAD(P)H oxidase activity, and ROS production were assessed. In addition, endothelial function, expression of angiotensin II (Ang II) receptor type 1 and 2 (AT1-R and AT2-R), and Ang II-mediated vasoconstriction were determined. ET resulted in a significant lower expression of gp91phox (23.1±0.5 versus 69.1±18.1 arbitrary units, training versus control), p22phox (0.7±0.3 versus 2.0±0.5 arbitrary units), and Nox4 (2.7±1.2 versus 5.4±1.0 arbitrary units). Enzymatic activity (2.1±0.3 versus 4.9±0.4 mU/mg) and ROS generation (0.02±0.01 versus 0.06±0.02 arbitrary units) were significantly lower in the training compared with the control group. On a functional level, ET resulted in improved acetylcholine-mediated vasodilatation and a 49% reduction in Ang II–induced vasoconstriction, accompanied by lower AT1-R (3.7±0.8 versus 16.6±5.7 arbitrary units, training versus control) and higher AT2-R (7.8±2.5 versus 1.6±0.7 arbitrary units) mRNA expression. Conclusions—ET reduces vascular expression of NAD(P)H oxidase and AT1-R, resulting in decreased local ROS generation. These molecular effects converge in a reduced Ang II–mediated vasoconstriction.


Circulation | 2005

Effects of Exercise and Ischemia on Mobilization and Functional Activation of Blood-Derived Progenitor Cells in Patients With Ischemic Syndromes Results of 3 Randomized Studies

Marcus Sandri; Volker Adams; Stephan Gielen; Axel Linke; Karsten Lenk; Nicolle Kränkel; Dominik Lenz; Sandra Erbs; Dierk Scheinert; Friedrich W. Mohr; Gerhard Schuler; Rainer Hambrecht

Background—Exercise training (ET) has been shown to improve regional perfusion in ischemic syndromes. This might be partially related to a regeneration of diseased endothelium by circulating progenitor cells (CPCs) or CPC-derived vasculogenesis. The aim of the present study was to determine whether ischemic stimuli during ET are required to promote CPC mobilization in patients with cardiovascular diseases. Methods and Results—Patients with peripheral arterial occlusive disease (PAOD) were randomized to 4 weeks of daily ischemic ET or control (group A). Successfully revascularized patients with PAOD were randomized to 4 weeks of daily nonischemic ET or control (group B). Patients with stable coronary artery disease were subjected to 4 weeks of subischemic ET or control (group C). At baseline and after 4 weeks, the number of KDR+/CD34+ CPCs was determined by fluorescence-activated cell sorting analysis. Levels of vascular endothelial growth factor (VEGF) were measured by ELISA. A Matrigel assay was used to quantify CPC integration into vascular structures. Expression of the homing factor CXCR4 was determined by reverse transcription-polymerase chain reaction. In group A only, ischemic ET increased VEGF levels by 310% (P<0.05 versus control) associated with an increase in CPCs by 440% (P<0.05 versus control), increased CXCR4 expression, and enhanced integration of CPCs into endothelial networks. In contrast, subischemic ET in groups B and C increased CXCR4 expression and CPC integration. Conclusions—In training programs, symptomatic tissue ischemia seems to be a prerequisite for CPC mobilization. However, ischemic and subischemic ET programs affect CXCR4 expression of CPCs, which might lead to an improved CPC integration into endothelial networks.


Circulation | 2010

Human Adult Vena Saphena Contains Perivascular Progenitor Cells Endowed With Clonogenic and Proangiogenic Potential

Paola Campagnolo; Daniela Cesselli; Ayman Al Haj Zen; Antonio Paolo Beltrami; Nicolle Kränkel; Rajesh Katare; Gianni D. Angelini; Costanza Emanueli; Paolo Madeddu

Background— Clinical trials in ischemic patients showed the safety and benefit of autologous bone marrow progenitor cell transplantation. Non–bone marrow progenitor cells with proangiogenic capacities have been described, yet they remain clinically unexploited owing to their scarcity, difficulty of access, and low ex vivo expansibility. We investigated the presence, antigenic profile, expansion capacity, and proangiogenic potential of progenitor cells from the saphenous vein of patients undergoing coronary artery bypass surgery. Methods and Results— CD34-positive cells, negative for the endothelial marker von Willebrand factor, were localized around adventitial vasa vasorum. After dissection of the vein from surrounding tissues and enzymatic digestion, CD34-positive/CD31-negative cells were isolated by selective culture, immunomagnetic beads, or fluorescence-assisted cell sorting. In the presence of serum, CD34-positive/CD31-negative cells gave rise to a highly proliferative population that expressed pericyte/mesenchymal antigens together with the stem cell marker Sox2 and showed clonogenic and multilineage differentiation capacities. We called this population “saphenous vein–derived progenitor cells” (SVPs). In culture, SVPs integrated into networks formed by endothelial cells and supported angiogenesis through paracrine mechanisms. Reciprocally, endothelial cell–released factors facilitated SVP migration. These interactive responses were inhibited by Tie-2 or platelet-derived growth factor-BB blockade. Intramuscular injection of SVPs in ischemic limbs of immunodeficient mice improved neovascularization and blood flow recovery. At 14 days after transplantation, proliferating SVPs were still detectable in the recipient muscles, where they established N-cadherin–mediated physical contact with the capillary endothelium. Conclusions— SVPs generated from human vein CD34-positive/CD31-negative progenitor cells might represent a new therapeutic tool for angiogenic therapy in ischemic patients.


Circulation Research | 2009

Human CD133 + Progenitor Cells Promote the Healing of Diabetic Ischemic Ulcers by Paracrine Stimulation of Angiogenesis and Activation of Wnt Signaling

Luciola S Barcelos; Cécile Duplàa; Nicolle Kränkel; Gallia Graiani; Gloria Invernici; Rajesh Katare; Mauro Siragusa; Marco Meloni; Ilaria Campesi; Manuela Monica; Andreas Simm; Paola Campagnolo; Giuseppe Mangialardi; Lara Stevanato; Giulio Alessandri; Costanza Emanueli; Paolo Madeddu

We evaluated the healing potential of human fetal aorta–derived CD133+ progenitor cells and their conditioned medium (CD133+ CCM) in a new model of ischemic diabetic ulcer. Streptozotocin-induced diabetic mice underwent bilateral limb ischemia and wounding. One wound was covered with collagen containing 2×104 CD133+ or CD133− cells or vehicle. The contralateral wound, covered with only collagen, served as control. Fetal CD133+ cells expressed high levels of wingless (Wnt) genes, which were downregulated following differentiation into CD133− cells along with upregulation of Wnt antagonists secreted frizzled-related protein (sFRP)-1, -3, and -4. CD133+ cells accelerated wound closure as compared with CD133− or vehicle and promoted angiogenesis through stimulation of endothelial cell proliferation, migration, and survival by paracrine effects. CD133+ cells secreted high levels of vascular endothelial growth factor (VEGF)-A and interleukin (IL)-8. Consistently, CD133+ CCM accelerated wound closure and reparative angiogenesis, with this action abrogated by coadministering the Wnt antagonist sFRP-1 or neutralizing antibodies against VEGF-A or IL-8. In vitro, these effects were recapitulated following exposure of high-glucose-primed human umbilical vein endothelial cells to CD133+ CCM, resulting in stimulation of migration, angiogenesis-like network formation and induction of Wnt expression. The promigratory and proangiogenic effect of CD133+ CCM was blunted by sFRP-1, as well as antibodies against VEGF-A or IL-8. CD133+ cells stimulate wound healing by paracrine mechanisms that activate Wnt signaling pathway in recipients. These preclinical findings open new perspectives for the cure of diabetic ulcers.


Immunity | 2013

Abnormal High-Density Lipoprotein Induces Endothelial Dysfunction via Activation of Toll-like Receptor-2

Thimoteus Speer; Lucia Rohrer; Przemyslaw Blyszczuk; Rukshana Shroff; Kira Kuschnerus; Nicolle Kränkel; Gabriela Kania; Stephen Zewinger; Alexander Akhmedov; Yi Shi; Tina Martin; Damir Perisa; Stephan Winnik; Maja Müller; Urban Sester; Gabriel Wernicke; Andreas Jung; Ursula Gutteck; Urs Eriksson; Jürgen Geisel; John Deanfield; Arnold von Eckardstein; Thomas F. Lüscher; Danilo Fliser; Ferdinand H. Bahlmann; Ulf Landmesser

Endothelial injury and dysfunction (ED) represent a link between cardiovascular risk factors promoting hypertension and atherosclerosis, the leading cause of death in Western populations. High-density lipoprotein (HDL) is considered antiatherogenic and known to prevent ED. Using HDL from children and adults with chronic kidney dysfunction (HDL(CKD)), a population with high cardiovascular risk, we have demonstrated that HDL(CKD) in contrast to HDL(Healthy) promoted endothelial superoxide production, substantially reduced nitric oxide (NO) bioavailability, and subsequently increased arterial blood pressure (ABP). We have identified symmetric dimethylarginine (SDMA) in HDL(CKD) that causes transformation from physiological HDL into an abnormal lipoprotein inducing ED. Furthermore, we report that HDL(CKD) reduced endothelial NO availability via toll-like receptor-2 (TLR-2), leading to impaired endothelial repair, increased proinflammatory activation, and ABP. These data demonstrate how SDMA can modify the HDL particle to mimic a damage-associated molecular pattern that activates TLR-2 via a TLR-1- or TLR-6-coreceptor-independent pathway, linking abnormal HDL to innate immunity, ED, and hypertension.


Circulation-heart Failure | 2010

Exercise Training in Patients with Advanced Chronic Heart Failure (NYHA IIIb) Promotes Restoration of Peripheral Vasomotor Function, Induction of Endogenous Regeneration, and Improvement of Left-Ventricular Function

Sandra Erbs; Robert Höllriegel; Axel Linke; Ephraim B Beck; Volker Adams; Stephan Gielen; Sven Möbius-Winkler; Marcus Sandri; Nicolle Kränkel; Rainer Hambrecht; Gerhard Schuler

Background—Attenuated peripheral perfusion in patients with advanced chronic heart failure (CHF) is partially the result of endothelial dysfunction. This has been causally linked to an impaired endogenous regenerative capacity of circulating progenitor cells (CPC). The aim of this study was to elucidate whether exercise training (ET) affects exercise intolerance and left ventricular (LV) performance in patients with advanced CHF (New York Heart Association class IIIb) and whether this is associated with correction of peripheral vasomotion and induction of endogenous regeneration. Methods and Results—Thirty-seven patients with CHF (LV ejection fraction 24±2%) were randomly assigned to 12 weeks of ET or sedentary lifestyle (control). At the beginning of the study and after 12 weeks, maximal oxygen consumption (Vo2max) and LV ejection fraction were determined; the number of CD34+/KDR+ CPCs was quantified by flow cytometry and CPC functional capacity was determined by migration assay. Flow-mediated dilation was assessed by ultrasound. Capillary density was measured in skeletal muscle tissue samples. In advanced CHF, ET improved Vo2max by +2.7±2.2 versus −0.8±3.1 mL/min/kg in control (P=0.009) and LV ejection fraction by +9.4±6.1 versus −0.8±5.2% in control (P<0.001). Flow-mediated dilation improved by +7.43±2.28 versus +0.09±2.18% in control (P<0.001). ET increased the number of CPC by +83±60 versus −6±109 cells/mL in control (P=0.014) and their migratory capacity by +224±263 versus −12±159 CPC/1000 plated CPC in control (P=0.03). Skeletal muscle capillary density increased by +0.22±0.10 versus −0.02±0.16 capillaries per fiber in control (P<0.001). Conclusions—Twelve weeks of ET in patients with advanced CHF is associated with augmented regenerative capacity of CPCs, enhanced flow-mediated dilation suggestive of improvement in endothelial function, skeletal muscle neovascularization, and improved LV function. Clinical Trial Registration—http://www.clinicaltrials.gov. Unique Identifier: NCT00176384.


Circulation Research | 2008

Role of kinin B2 receptor signaling in the recruitment of circulating progenitor cells with neovascularization potential

Nicolle Kränkel; Rajesh Katare; Mauro Siragusa; Luciola S Barcelos; Paola Campagnolo; Giuseppe Mangialardi; Orazio Fortunato; Gaia Spinetti; Nguyen Tran; Kai Zacharowski; Wojciech Wojakowski; Iwona Mroz; Andrew Herman; Jocelyn E. Manning Fox; Patrick E. MacDonald; Joost P. Schanstra; Jean Loup Bascands; Raimondo Ascione; Gianni D. Angelini; Costanza Emanueli; Paolo Madeddu

Reduced migratory function of circulating angiogenic progenitor cells (CPCs) has been associated with impaired neovascularization in patients with cardiovascular disease (CVD). Previous findings underline the role of the kallikrein-kinin system in angiogenesis. We now demonstrate the involvement of the kinin B2 receptor (B2R) in the recruitment of CPCs to sites of ischemia and in their proangiogenic action. In healthy subjects, B2R was abundantly present on CD133+ and CD34+ CPCs as well as cultured endothelial progenitor cells (EPCs) derived from blood mononuclear cells (MNCs), whereas kinin B1 receptor expression was barely detectable. In transwell migration assays, bradykinin (BK) exerts a potent chemoattractant activity on CD133+ and CD34+ CPCs and EPCs via a B2R/phosphoinositide 3-kinase/eNOS-mediated mechanism. Migration toward BK was able to attract an MNC subpopulation enriched in CPCs with in vitro proangiogenic activity, as assessed by Matrigel assay. CPCs from cardiovascular disease patients showed low B2R levels and decreased migratory capacity toward BK. When injected systemically into wild-type mice with unilateral limb ischemia, bone marrow MNCs from syngenic B2R-deficient mice resulted in reduced homing of sca-1+ and cKit+flk1+ progenitors to ischemic muscles, impaired reparative neovascularization, and delayed perfusion recovery as compared with wild-type MNCs. Similarly, blockade of the B2R by systemic administration of icatibant prevented the beneficial effect of bone marrow MNC transplantation. BK-induced migration represents a novel mechanism mediating homing of circulating angiogenic progenitors. Reduction of BK sensitivity in progenitor cells from cardiovascular disease patients might contribute to impaired neovascularization after ischemic complications.


Circulation | 2012

Loss of AngiomiR-126 and 130a in Angiogenic Early Outgrowth Cells From Patients With Chronic Heart Failure Role for Impaired In Vivo Neovascularization and Cardiac Repair Capacity

Philipp Jakob; Carola Doerries; Sylvie Briand; Pavani Mocharla; Nicolle Kränkel; Christian Besler; Maja Mueller; Costantina Manes; Christian Templin; Christof Baltes; Markus Rudin; Heiner Adams; Mathias Wolfrum; Georg Noll; Frank Ruschitzka; Thomas F. Lüscher; Ulf Landmesser

Background— MicroRNAs are key regulators of angiogenic processes. Administration of angiogenic early outgrowth cells (EOCs) or CD34+ cells has been suggested to improve cardiac function after ischemic injury, in particular by promoting neovascularization. The present study therefore examines regulation of angiomiRs, microRNAs involved in angiogenesis, in angiogenic EOCs and circulating CD34+ cells from patients with chronic heart failure (CHF) and the role for their cardiac repair capacity. Methods and Results— Angiogenic EOCs and CD34+ cells were isolated from patients with CHF caused by ischemic cardiomyopathy (n=45) and healthy subjects (n=35). In flow cytometry analyses, angiogenic EOCs were largely myeloid and positive for alternatively activated M2 macrophage markers. In vivo cardiac neovascularization and functional repair capacity were examined after transplantation into nude mice with myocardial infarction. Cardiac transplantation of angiogenic EOCs from healthy subjects markedly increased neovascularization and improved cardiac function, whereas no such effect was observed after transplantation of angiogenic EOCs from patients with CHF. Real-time polymerase chain reaction analysis of 14 candidate angiomiRs, expressed in angiogenic EOCs, revealed a pronounced loss of angiomiR-126 and -130a in angiogenic EOCs from patients with CHF that was also observed in circulating CD34+ cells. Anti–miR-126 transfection markedly impaired the capacity of angiogenic EOCs from healthy subjects to improve cardiac function. miR-126 mimic transfection increased the capacity of angiogenic EOCs from patients with CHF to improve cardiac neovascularization and function. Conclusions— The present study reveals a loss of angiomiR-126 and -130a in angiogenic EOCs and circulating CD34+ cells from patients with CHF. Reduced miR-126 expression was identified as a novel mechanism limiting their capacity to improve cardiac neovascularization and function that can be targeted by miR-126 mimic transfection.Background— MicroRNAs are key regulators of angiogenic processes. Administration of angiogenic early outgrowth cells (EOCs) or CD34+ cells has been suggested to improve cardiac function after ischemic injury, in particular by promoting neovascularization. The present study therefore examines regulation of angiomiRs, microRNAs involved in angiogenesis, in angiogenic EOCs and circulating CD34+ cells from patients with chronic heart failure (CHF) and the role for their cardiac repair capacity. Methods and Results— Angiogenic EOCs and CD34+ cells were isolated from patients with CHF caused by ischemic cardiomyopathy (n=45) and healthy subjects (n=35). In flow cytometry analyses, angiogenic EOCs were largely myeloid and positive for alternatively activated M2 macrophage markers. In vivo cardiac neovascularization and functional repair capacity were examined after transplantation into nude mice with myocardial infarction. Cardiac transplantation of angiogenic EOCs from healthy subjects markedly increased neovascularization and improved cardiac function, whereas no such effect was observed after transplantation of angiogenic EOCs from patients with CHF. Real-time polymerase chain reaction analysis of 14 candidate angiomiRs, expressed in angiogenic EOCs, revealed a pronounced loss of angiomiR-126 and -130a in angiogenic EOCs from patients with CHF that was also observed in circulating CD34+ cells. Anti–miR-126 transfection markedly impaired the capacity of angiogenic EOCs from healthy subjects to improve cardiac function. miR-126 mimic transfection increased the capacity of angiogenic EOCs from patients with CHF to improve cardiac neovascularization and function. Conclusions— The present study reveals a loss of angiomiR-126 and -130a in angiogenic EOCs and circulating CD34+ cells from patients with CHF. Reduced miR-126 expression was identified as a novel mechanism limiting their capacity to improve cardiac neovascularization and function that can be targeted by miR-126 mimic transfection. # Clinical Perspective {#article-title-54}


Circulation Research | 2011

Tissue kallikrein is essential for invasive capacity of circulating proangiogenic cells.

Gaia Spinetti; Orazio Fortunato; Daniela Cordella; Paola Portararo; Nicolle Kränkel; Rajesh Katare; Graciela B. Sala-Newby; Christine Richer; Marie Vincent; François Alhenc-Gelas; Giancarlo Tonolo; Sara Cherchi; Costanza Emanueli; Paolo Madeddu

Rationale: Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. Objective: To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. Methods and Results: Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow–derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B2 receptor (B2R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B2R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B2R expression rescued the diabetic phenotype. Conclusions: This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor–dependent mechanisms.


European Journal of Preventive Cardiology | 2008

Circulating progenitor cells decrease immediately after marathon race in advanced-age marathon runners

Volker Adams; Axel Linke; Frank Breuckmann; Kirsten Leineweber; Sandra Erbs; Nicolle Kränkel; Martina Bröcker-Preuss; Felix Woitek; Raimun d Erbel; Gerd Heusch; Rai ner Hambrecht; Gerhard Schuler; Stefan Möhlenkamp

Introduction Exercise is thought to stimulate the release of hematopoietic and endothelial progenitor cells (EPC) from the bone marrow. Little is known about the influence of strenuous exercise on the content of circulating progenitor cells. The aim of this study was to investigate the influence of a marathon race on the amount of circulating progenitor cells immediately after the race in advanced-aged runners. Methods Sixty-eight healthy marathon runners (age: 57 ± 6 years) were included in this study. Blood cell counts were evaluated by standard methods, and circulating progenitor cells before and immediately after the race were quantified by fluorescence-activated cell sorter (FACS). Vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF) was quantified by enzyme-linked immunosorbent assay. Results A marathon race led to a significant increase in white blood cell count (5283 ± 155 vs. 13706 ± 373 cells/μl; P < 0.001). Fluorescence-activated cell sorter analysis revealed a significant decrease of CD34pos cells (1829 ± 115 vs. 1175 ± 75 cells/ml blood; P < 0.0001), CD117pos cells (2478 ± 245 vs. 2193 ± 85 cells/ml blood; P < 0.05), and CD133pos cells (3505 ± 286 vs. 2239 ± 163 cells/ml blood; P < 0.001). No significant change was observed for EPCs defined as CD34pos/VEGF-R2pos cells (117 ± 8 vs. 128 ± 9cells/ml blood; P = 0.33). With respect to VEGF a significant downregulation was evident directly after the race (48.9 ± 8.0 vs. 34.0 ± 7.5 pg/ml; P < 0.05), whereas no change was obvious in EGF levels. Conclusion The results of our study suggest that finishing a marathon race will lead to an inflammatory response and downregulation of circulating hematopoietic stem cells. With respect to EPCs no change is observed, which may be because of a greater differentiation of the remaining CD34pos cells towards EPCs.

Collaboration


Dive into the Nicolle Kränkel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge