Network


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

Hotspot


Dive into the research topics where Claudia Schrimpf is active.

Publication


Featured researches published by Claudia Schrimpf.


Cytokine | 2011

Acute administration of recombinant Angiopoietin-1 ameliorates multiple-organ dysfunction syndrome and improves survival in murine sepsis

Sascha David; Joon-Keun Park; Matijs van Meurs; Jan G. Zijlstra; Christian Koenecke; Claudia Schrimpf; Nelli Shushakova; Faikah Gueler; Hermann Haller; Philipp Kümpers

INTRODUCTION Endothelial activation leading to vascular barrier breakdown plays an essential role in the pathophysiology of multiple-organ dysfunction syndrome (MODS) in sepsis. Increasing evidence suggests that the function of the vessel-protective factor Angiopoietin-1 (Ang-1), a ligand of the endothelial-specific Tie2 receptor, is inhibited by its antagonist Angiopoietin-2 (Ang-2) during sepsis. In order to reverse the effects of the sepsis-induced suppression of Ang-1 and elevation of Ang-2 we aimed to investigate whether an intravenous injection of recombinant human (rh) Ang-1 protects against MODS in murine sepsis. METHODS Polymicrobiological abdominal sepsis was induced by cecal ligation and puncture (CLP). Mice were treated with either 1 μg of intravenous rhAng-1 or control buffer immediately after CLP induction and every 8h thereafter. Sham-operated animals served as time-matched controls. RESULTS Compared to buffer-treated controls, rhAng-1 treated septic mice showed significant improvements in several hematologic and biochemical indicators of MODS. Moreover, rhAng-1 stabilized endothelial barrier function, as evidenced by inhibition of protein leakage from lung capillaries into the alveolar compartment. Histological analysis revealed that rhAng-1 treatment attenuated leukocyte infiltration in lungs and kidneys of septic mice, probably due to reduced endothelial adhesion molecule expression in rhAng-1 treated mice. Finally, the protective effects of rhAng-1 treatment were reflected by an improved survival time in a lethal CLP model. CONCLUSIONS In a clinically relevant murine sepsis model, intravenous rhAng-1 treatment alone is sufficient to significantly improve a variety of sepsis-associated organ dysfunctions and survival time, most likely by preserving endothelial barrier function. Further studies are needed to pave the road for clinical application of this therapy concept.


Journal of Vascular Research | 2014

The Role of Pericyte Detachment in Vascular Rarefaction

Claudia Schrimpf; Omke E. Teebken; Mathias Wilhelmi; Jeremy S. Duffield

Background: Pericytes surround endothelial cells at the perivascular interface. Signaling between endothelial cells and pericytes is crucial for capillary homeostasis, as pericytes stabilize vessels and regulate many microvascular functions. Recently it has been shown that pericytes are able to detach from the vascular wall and contribute to fibrosis by becoming scar-forming myofibroblasts in many organs including the kidney. At the same time, the loss of pericytes within the perivascular compartment results in vulnerable capillaries which are prone to instability, pathological angiogenesis, and, ultimately, rarefaction. Aims: This review will give an overview of pericyte-endothelial cell interactions, summarize the signaling pathways that have been identified to be involved in pericyte detachment from the vascular wall, and present pathological endothelial responses in the context of disease of the kidney.


Critical Care Medicine | 2015

Drug Repurposing Screen Identifies Foxo1-Dependent Angiopoietin-2 Regulation in Sepsis.

Chandra C. Ghosh; Kristina Thamm; Anthony V. Berghelli; Claudia Schrimpf; Manish R. Maski; Tanaz Abid; Katelyn Milam; Augustine Rajakumar; Ansgar Santel; Jan T. Kielstein; Asif Ahmed; David R Thickett; Keqin Wang; Maureen Chase; Michael W. Donnino; William C. Aird; Hermann Haller; Sascha David; Samir M. Parikh

Objective:The recent withdrawal of a targeted sepsis therapy has diminished pharmaceutical enthusiasm for developing novel drugs for the treatment of sepsis. Angiopoietin-2 is an endothelial-derived protein that potentiates vascular inflammation and leakage and may be involved in sepsis pathogenesis. We screened approved compounds for putative inhibitors of angiopoietin-2 production and investigated underlying molecular mechanisms. Design:Laboratory and animal research plus prospective placebo-controlled randomized controlled trial (NCT00529139) and retrospective analysis (NCT00676897). Setting:Research laboratories of Hannover Medical School and Harvard Medical School. Patients:Septic patients/C57Bl/6 mice and human endothelial cells. Interventions:Food and Drug Administration–approved library screening. Measurements and Main Results:In a cell-based screen of more than 650 Food and Drug Administration–approved compounds, we identified multiple members of the 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitor drug class (referred to as statins) that suppressed angiopoietin-2. Simvastatin inhibited 3-hydroxy-3-methyl-glutaryl-CoA reductase, which in turn activated PI3K-kinase. Downstream of this signaling, PI3K-dependent phosphorylation of the transcription factor Foxo1 at key amino acids inhibited its ability to shuttle to the nucleus and bind cis-elements in the angiopoietin-2 promoter. In septic mice, transient inhibition of angiopoietin-2 expression by liposomal siRNA in vivo improved absolute survival by 50%. Simvastatin had a similar effect, but the combination of angiopoietin-2 siRNA and simvastatin showed no additive benefit. To verify the link between statins and angiopoietin-2 in humans, we performed a pilot matched case-control study and a small randomized placebo-controlled trial demonstrating beneficial effects on angiopoietin-2. Conclusions:3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors may operate through a novel Foxo1-angiopoietin-2 mechanism to suppress de novo production of angiopoietin-2 and thereby ameliorate manifestations of sepsis. Given angiopoietin-2’s dual role as a biomarker and candidate disease mediator, early serum angiopoietin-2 measurement may serve as a stratification tool for future trials of drugs targeting vascular leakage.


PLOS ONE | 2015

Renal function interferes with copeptin in prediction of major adverse cardiac events in patients undergoing vascular surgery.

Claudia Schrimpf; Hans-Joerg Gillmann; Bianca Sahlmann; Antje Meinders; Jan Larmann; Mathias Wilhelmi; Thomas Aper; Saad Rustum; Ralf Lichtinghagen; Gregor Theilmeier; Omke E. Teebken

OBJECTIVE Precise perioperative risk stratification is important in vascular surgery patients who are at high risk for major adverse cardiovascular events (MACE) peri- and postoperatively. In clinical practice, the patients perioperative risk is predicted by various indicators, e.g. revised cardiac index (RCRI) or modifications thereof. Patients suffering from chronic kidney disease (CKD) are stratified into a higher risk category. We hypothesized that Copeptin as a novel biomarker for hemodynamic stress could help to improve the prediction of perioperative cardiovascular events in patients undergoing vascular surgery including patients with chronic kidney disease. METHODS 477 consecutive patients undergoing abdominal aortic, peripheral arterial or carotid surgery from June 2007 to October 2012 were prospectively enrolled. Primary endpoint was 30-day postoperative major adverse cardiovascular events (MACE). RESULTS 41 patients reached the primary endpoint, including 63.4% aortic, 26.8% carotid, and 9.8% peripheral surgeries. Linear regression analysis showed that RCRI (P< .001), pre- (P< .001), postoperative Copeptin (P< .001) and Copeptin level change (P= .001) were associated with perioperative MACE, but CKD remained independently associated with MACE and Copeptin levels. Multivariate regression showed that increased Copeptin levels added risk predictive information to the RCRI (P= .003). Especially in the intermediate RCRI categories was Copeptin significantly associated with the occurrence of MACE. (P< .05 Kruskal Wallis test). Subdivision of the study cohort into CKD stages revealed that preoperative Copeptin was significantly associated with CKD stages (P< .0001) and preoperative Copeptin measurements could not predict MACE in patients with more severe CKD stages. CONCLUSION Preoperative Copeptin loses its risk predictive potential for perioperative MACE in patients with chronic kidney disease undergoing vascular surgery.


BioNanoMaterials | 2015

Evaluation of autologous tissue sources for the isolation of endothelial cells and adipose tissue-derived mesenchymal stem cells to pre-vascularize tissue-engineered vascular grafts

Skadi Lau; Claudia Schrimpf; Melanie Klingenberg; Fabian Helfritz; Thomas Aper; Axel Haverich; Mathias Wilhelmi; Ulrike Böer

Abstract Currently used synthetic vascular grafts bear a high infection risk due to insufficient microvascularization of the graft wall disabling the infiltration of immune cells. Tissue-engineered grafts with a functional pre-vascularization thus would be desirable. However, autologous tissue sources for capillary forming cells need to be evaluated. Here, peripheral blood outgrowth endothelial cells (PB-OEC) from 17 healthy donors and pericyte-like mesenchymal stem cells derived from adipose tissue (ASC) of 17 patients scheduled for visceral surgery were characterized and investigated regarding their ability to form capillary-like networks in plasma-derived fibrin gels. To obtain proliferating PB-OEC with endothelial cell-specific properties (CD31-, VE-cadherin-expression, ac-LDL uptake and three-dimensional (3D)-tube formation in fibrin gels) both enrichment of CD34+ blood cells and young donor age was necessary (7/17, age≤24 years). In contrast, all isolated ASC revealed the expression of surface antigens expressed on pericytes [human neural/glial antigen-2 (hNG2), platelet-derived growth factor receptor β (PDGF-Rβ)] and showed mesodermal differentiation capacity. Moreover, co-culture of PB-OEC and ASC in fibrin gels resulted in highly branched capillary-like networks with significantly increased tube length (2.9-fold, p<0.0001) and number of junctions (8-fold, p<0.0001). In conclusion, successful cell isolation from autologous tissues for pre-vascularization of vascular grafts has been demonstrated although certain limitations for autologous EC require further strategies to enable the use of allogeneic cells.


Journal of Intensive Care Medicine | 2017

Adrenomedullin Is Associated With Surgical Trauma and Impaired Renal Function in Vascular Surgery Patients.

Hans-Jörg Gillmann; Antje Meinders; Jan Larmann; Bianca Sahlmann; Claudia Schrimpf; Thomas Aper; Ralf Lichtinghagen; Omke E. Teebken; Gregor Theilmeier

Background: Patients undergoing vascular surgery are prone to perioperative organ injury because of both higher prevalence of cardiovascular risk factors and the extent of surgery. Early detection of organ failure is essential to facilitate appropriate medical care. Midregional pro-adrenomedullin (MR-proADM) has been investigated in acute medical care settings to guide clinical decision-making regarding patient pathways and to identify patients prone to imminent cardiovascular or inflammatory complications. In this study, we evaluated the impact of perioperative MR-proADM levels as an early marker of perioperative cardiovascular and inflammatory stress reactions and kidney injury. Methods: The study was conducted as a monocentric, prospective, noninterventional trial at Hannover Medical School, Germany. A total of 454 consecutive patients who underwent open vascular surgery were followed from the day prior to until 30 days after surgery. The composite primary end point was defined as the occurrence of major adverse cardiac events (MACEs), acute kidney injury (AKI), or systemic inflammatory response syndrome (SIRS). Measurements were correlated with both medical history and postoperative MACE, AKI, or SIRS using univariate and multivariate regression analysis. Results: One hundred thirty-nine (31%) of the patients reached the primary end point within the study interval. Midregional pro-adrenomedullin change was associated with the combined primary end point and with the intensity of surgical trauma. Midregional pro-adrenomedullin change was increased in patients reaching the secondary end points, SIRS (optimal cutoff: 0.2 nmol/L) and AKI (optimal cutoff: 0.7 nmol/L), but not in patients with MACEs. Conclusion: Increased levels of MR-proADM within the perioperative setting (1) were linked to the invasiveness of surgery and (2) identified patients with ongoing loss of renal function. Increased MR-proADM levels may therefore identify a subgroup of patients prone to excessive cardiovascular stress but did not directly correlate with adverse cardiac events. Consistently low levels of MR-proADM may identify a subgroup of patients with acceptable low risk to guide discharge from high-density care units.


The Annals of Thoracic Surgery | 2015

Thoracic Endovascular Aortic Repair After Iatrogenic Aortic Dissection and False Lumen Stent Grafting

Claudia Schrimpf; Omke E. Teebken; Mathias Wilhelmi

Iatrogenic aortic dissections are a severe complication after thoracic endovascular aortic repair, and treatment guidelines do not exist. Herein, we report a patient who experienced an iatrogenic type B aortic dissection during elective thoracic endovascular aortic repair and suggest an interventional treatment option.


The Thoracic & Cardiovascular Surgeon Reports | 2013

Unusual Case of an Arterial Thoracic Outlet Syndrome due to Srb Anomaly

Saad Rustum; Thomas Rodt; Omke E. Teebken; Claudia Schrimpf; Thomas Aper; Mathias Wilhelmi

A thoracic outlet syndrome (TOS) is caused by arterial or nervous obstruction because of skeletal or muscular anomalies and hypertrophies. Congenital rib anomalies occur with low incidences (0.15–0.31%), predominantly affect the right side and are normally diagnosed at a young age or remain asymptomatic throughout life. Here, we report on the unusual case of a 71-year-old female patient with subacute ischemia of the left arm due to a TOS resulting from Srb anomaly, a very rare congenital rib anomaly.


Tissue Engineering Part A | 2017

Low immunogenic endothelial cells maintain morphofunctional properties needed for tissue engineering

Skadi Lau; Dorothee Eicke; Marco Carvalho Oliveira; Bettina Wiegmann; Claudia Schrimpf; Axel Haverich; Rainer Blasczyk; Mathias Wilhelmi; Constanςa Figueiredo; Ulrike Böer

The limited availability of native vessels suitable for the application as hemodialysis shunts or bypass material demands new strategies in cardiovascular surgery. Tissue-engineered vascular grafts containing autologous cells are considered ideal vessel replacements due to the low risk of rejection. However, endothelial cells (EC), which are central components of natural blood vessels, are difficult to obtain from elderly patients of poor health. Umbilical cord blood represents a promising alternative source for EC, but their allogeneic origin corresponds with the risk of rejection after allotransplantation. To reduce this risk, the human leukocyte antigen class I (HLA I) complex was stably silenced by lentiviral vector-mediated RNA interference (RNAi) in EC from peripheral blood and umbilical cord blood and vein. EC from all three sources were transduced by 93.1% ± 4.8% and effectively, HLA I-silenced by up to 67% compared to nontransduced (NT) cells or transduced with a nonspecific short hairpin RNA, respectively. Silenced EC remained capable to express characteristic endothelial surface markers such as CD31 and vascular endothelial cadherin important for constructing a tight barrier, as well as von Willebrand factor and endothelial nitric oxide synthase important for blood coagulation and vessel tone regulation. Moreover, HLA I-silenced EC were still able to align under unidirectional flow, to take up acetylated low-density lipoprotein, and to form capillary-like tube structures in three-dimensional fibrin gels similar to NT cells. In particular, addition of adipose tissue-derived mesenchymal stem cells significantly improved tube formation capability of HLA I-silenced EC toward long and widely branched vascular networks necessary for prevascularizing vascular grafts. Thus, silencing HLA I by RNAi represents a promising technique to reduce the immunogenic potential of EC from three different sources without interfering with EC-specific morphological and functional properties required for vascular tissue engineering. This extends the spectrum of available cell sources from autologous to allogeneic sources, thereby accelerating the generation of tissue-engineered vascular grafts in acute clinical cases.


Journal of Tissue Engineering | 2017

Outgrowing endothelial and smooth muscle cells for tissue engineering approaches

Moritz Kolster; Mathias Wilhelmi; Claudia Schrimpf; Andres Hilfiker; Axel Haverich; Thomas Aper

In recent years, circulating progenitors of endothelial cells and smooth muscle cells were identified in the peripheral blood. In our study, we evaluated the utilization of both cell types isolated and differentiated from peripheral porcine blood in terms for their use for tissue engineering purposes. By means of density gradient centrifugation, the monocyte fraction from porcine blood was separated, split, and cultivated with specific culture media with either endothelial cell growth medium-2 or smooth muscle cell growth medium-2 for the differentiation of endothelial cells or smooth muscle cells. Obtained cells were characterized at an early stage of cultivation before the first passage and a late stage (fourth passage) on the basis of the expression of the antigens CD31, CD34, CD45, nitric oxide synthase, and the contractile filaments smooth-muscle alpha-actin (sm-alpha-actin) and smoothelin. Functional characterization was done based on the secretion of nitric oxide, the formation of a coherent monolayer on polytetrafluoroethylene, and capillary sprouting. During cultivation in both endothelial cell growth medium-2 and smooth muscle cell growth medium-2, substantially two types of cells grew out: early outgrown CD45-positive cells, which disappeared during further cultivation, and in 85% (n = 17/20) of cultures cultivated with endothelial cell growth medium-2 colony-forming late outgrowth endothelial cells. During cultivation with smooth muscle cell growth medium-2 in 80% (n = 16/20) of isolations colony-forming late outgrowth smooth muscle cells entered the stage. Cultivation with either endothelial cell growth medium-2 or smooth muscle cell growth medium-2 had selective effect on the late outgrown cells to that effect that the number of CD31-positive cells increased from 34.8% ± 13% to 83.9% ± 8% in cultures cultivated with endothelial cell growth medium-2 and the number of sm-α-actin+ cells increased from 52.6% ± 18% to 88% ± 5% in cultures cultivated with smooth muscle cell growth medium-2, respectively. Functional analyses revealed significantly higher levels of nitric oxide secretion, endothelialization capacity, and capillary formation in not expanded cultures cultivated with endothelial cell growth medium-2 in comparison to later stages of cultivation and mature aortic cells. Blood seems to be a reliable and feasible source for the isolation of both endothelial and smooth muscle cells for application in tissue engineering approaches. Whereas, early co-cultures of early and late outgrowth cells provide functional advantages, the differentiation of cells can be directed selectively by the used culture medium for the expansion of highly proliferative late outgrowth endothelial cells and late outgrowth smooth muscle cells, respectively.

Collaboration


Dive into the Claudia Schrimpf's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Saad Rustum

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Thomas Aper

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sascha David

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar

Ulrike Böer

Hannover Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Larmann

Hannover Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge