Network


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

Hotspot


Dive into the research topics where Francis M. Wulfert is active.

Publication


Featured researches published by Francis M. Wulfert.


Shock | 2007

Early organ-specific endothelial activation during hemorrhagic shock and resuscitation

Matijs van Meurs; Francis M. Wulfert; Ageeth J. Knol; Ann De Haes; Martin C. Houwertjes; Leon Aarts; Grietje Molema

Multiple organ dysfunction syndrome (MODS) is a complication of hemorrhagic shock (HS) and related to high morbidity and mortality. Interaction of activated neutrophils and endothelial cells is considered to play a prominent role in the pathophysiology of MODS. Insight in the nature and molecular basis of endothelial cell activation during HS can assist in identifying new rational targets for early therapeutic intervention. In this study, we examined the kinetics and organ specificity of endothelial cell activation in a mouse model of HS. Anesthetized male mice were subjected to controlled hemorrhage to a MAP of 30 mmHg. Mice were killed after 15, 30, 60, or 90 min of HS. After 90 min of hemorrhagic shock, a group of mice was resuscitated with 6% hydroxyethyl starch 130/0.4. Untreated mice and sham shock mice that underwent instrumentation and 90 min of anesthesia without shock served as controls. Gene expression levels of inflammatory endothelial cell activation (P-selectin, E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1) and hypoxia-responsive genes (vascular endothelial growth factor and hypoxia-inducible factor 1&agr;) were quantified in kidney, liver, lung, brain, and heart tissue by quantitative reverse-transcription-polymerase chain reaction. Furthermore, we examined a selection of these genes with regard to protein expression and localization using immunohistochemical analysis. Induction of inflammatory genes occurred early during HS and already before resuscitation. Expression of adhesion molecules was significantly induced in all organs, albeit to a different extent depending on the organ. Endothelial genes CD31 and VE-cadherin, which function in endothelial cell homeostasis and integrity, were not affected during the shock phase except for VE-cadherin in the liver, which showed increased mRNA levels. The rapid inflammatory activation was not paralleled by induction of hypoxia-responsive genes. This study demonstrated the occurrence of early and organ-specific endothelial cell activation during hemorrhagic shock, as presented by induced expression of inflammatory genes. This implies that early therapeutic intervention at the microvascular level may be a rational strategy to attenuate MODS.


American Journal of Physiology-renal Physiology | 2009

Shock-induced stress induces loss of microvascular endothelial Tie2 in the kidney which is not associated with reduced glomerular barrier function

Matijs van Meurs; Neng F. Kurniati; Francis M. Wulfert; Sigridur A. Ásgeirsdóttir; Inge A. M. de Graaf; Simon C. Satchell; Peter W. Mathieson; Rianne M. Jongman; Philipp Kümpers; Jan G. Zijlstra; Peter Heeringa; Grietje Molema

Both hemorrhagic shock and endotoxemia induce a pronounced vascular activation in the kidney which coincides with albuminuria and glomerular barrier dysfunction. We hypothesized that changes in Tie2, a vascular restricted receptor tyrosine kinase shown to control microvascular integrity and endothelial inflammation, underlie this loss of glomerular barrier function. In healthy murine and human kidney, Tie2 is heterogeneously expressed in all microvascular beds, although to different extents. In mice subjected to hemorrhagic and septic shock, Tie2 mRNA and protein were rapidly, and temporarily, lost from the renal microvasculature, and normalized within 24 h after initiation of the shock insult. The loss of Tie2 protein could not be attributed to shedding as both in mice and healthy volunteers subjected to endotoxemia, sTie2 levels in the systemic circulation did not change. In an attempt to identify the molecular control of Tie2, we activated glomerular endothelial cell cultures and human kidney slices in vitro with LPS or TNF-alpha, but did not observe a change in Tie2 mRNA levels. In parallel to the loss of Tie2 in vivo, an overt influx of neutrophils in the glomerular compartment, which coincided with proteinuria, was seen. As neutrophil-endothelial cell interactions may play a role in endothelial adaptation to shock, and these effects cannot be mimicked in vitro, we depleted neutrophils before shock induction. While this neutrophil depletion abolished proteinuria, Tie2 was not rescued, implying that Tie2 may not be a major factor controlling maintenance of the glomerular filtration barrier in this model.


Anesthesiology | 2009

Adjunct Nitrous Oxide Normalizes Vascular Reactivity Changes after Hemorrhagic Shock in Mice under Isoflurane Anesthesia

Iryna V. Samarska; Matijs van Meurs; Hendrik Buikema; Martin C. Houwertjes; Francis M. Wulfert; Grietje Molema; Anne H. Epema; Robert H. Henning

Background:Hemorrhagic shock is associated with changes in vascular responsiveness that may lead to organ dysfunction and, ultimately, multiple organ dysfunction syndrome. Volatile anesthetics interfere with vasoresponsiveness, which may contribute to organ hypoperfusion. In this study, the authors examined the influence of adjunct nitrous oxide on the vascular responsiveness after short-term hemorrhagic shock under isoflurane anesthesia. Methods:Spontaneously breathing mice (n = 31, 27.6 ± 0.31 g) were anesthetized with isoflurane (1.4%) or with isoflurane (1.4%) and adjunct nitrous oxide (66%). Both groups were divided into Sham, Shock, and Resuscitated groups. Vascular reactivity to phenylephrine and acetylcholine and expression of cyclooxygenases were studied in the aorta. Results:In the isoflurane-anesthetized groups, the contractile response to phenylephrine was increased in the Shock as compared with the Sham and Resuscitated groups (Emax = 3.2 ± 0.4, 1.2 ± 0.4, and 2.5 ± 0.5 mN, respectively). Adjunct nitrous oxide increased phenylephrine contraction to a similar level in all three groups. In the Sham isoflurane group, acetylcholine caused a biphasic response: An initial relaxation followed by a contractile response sensitive to cyclooxygenases inhibition by indomethacine. The contractile response was abrogated in the isoflurane-anesthetized groups that underwent shock. In all groups, adjunct nitrous oxide preserved the contractile phase. Shock induced a down-regulation of cyclooxygenases-1, which was normalized by adjunct nitrous oxide. Conclusion:Adjunct nitrous oxide attenuates shock-induced changes in vascular reactivity and cyclooxygenases expression of mice under isoflurane anesthesia. This implies that vascular reactive properties during anesthesia in hemorrhagic shock conditions may be influenced by the choice of anesthetics.


Anesthesiology | 2012

Age-dependent role of microvascular endothelial and polymorphonuclear cells in lipopolysaccharide-induced acute kidney injury.

Francis M. Wulfert; Matijs van Meurs; Neng F. Kurniati; Rianne M. Jongman; Martin C. Houwertjes; Peter Heeringa; Michel Struys; Jan G. Zijlstra; Grietje Molema

Background: The incidence of acute kidney injury following severe sepsis is higher in the elderly. We hypothesized that microvascular endothelium is “primed” by aging and that sepsis represents a “second hit,” resulting in more severe microvascular complications. Methods: Three- and 18-months-old mice were intraperitoneally injected with 1,500 EU/g body weight lipopolysaccharide and sacrificed after 8 h. Flow cytometry and myeloperoxidase ELISA determined neutrophils in plasma. Quantitative reverse transcription polymerase chain reaction was used to analyze messenger ribonucleic acid levels of cell adhesion molecules P-selectin and E-selectin, vascular cell adhesion protein-1, intercellular adhesion molecule-1, angiopoietin receptor TIE-2, and angiopoietins Ang1 and Ang2. In kidney tissue we assessed neutrophil influx and E-selectin protein expression. Neutrophils were depleted with the monoclonal antibody NIMP. Results: At basal conditions, microvascular endothelial cell activation status was similar in both groups, except for a higher Ang-2 expression (P < 0.05) in the kidney of aged mice. Lipopolysaccharide-induced increase in neutrophil count was higher in old (3.3-fold change) compared with young mice (2.2-fold change). Messenger ribonucleic acid analysis showed higher upregulation of P- and E-selectin (P = 0.0004, P = 0.0007) after lipopolysaccharide administration in kidneys of elderly mice, which was confirmed at the protein level for E-selectin. Renal neutrophil influx in lipopolysaccharide-treated aged mice was increased (2.5-fold induction in aged and 2.1-fold in young, P < 0.0001). Polymorphonuclear cell depletion exaggerated the lipopolysaccharide-induced kidney injury. Conclusion: Ang-2 is increased in older mice, which might cause priming of the endothelial cells. Endothelium responded by a more extensive increase in expression of P- and E-selectin in older mice and increased polymorphonuclear cell influx.


Anesthesiology | 2011

Hemorrhagic Shock-induced Endothelial Cell Activation in a Spontaneous Breathing and a Mechanical Ventilation Hemorrhagic Shock Model Is Induced by a Proinflammatory Response and Not by Hypoxia

Matijs van Meurs; Francis M. Wulfert; Rianne M. Jongman; Martin Schipper; Martin C. Houwertjes; Michiel Vaneker; Gert Jan Scheffer; Luc J. Teppema; Leon Aarts; Peter Heeringa; Jan G. Zijlstra; Grietje Molema

Introduction: The interaction between neutrophils and activated endothelium is essential for the development of multiple organ dysfunction in patients with hemorrhagic shock (HS). Mechanical ventilation frequently is used in patients with HS. The authors sought to investigate the consequences of mechanical ventilation of mice subjected to HS on microvascular endothelial activation in the lung and kidney. Methods: Anesthetized wild type C57BL/6 male mice were subjected to controlled hemorrhage; subgroups of mice were mechanically ventilated during the HS insult. To study the effect of acute hypoxia on the mice, the animals were housed in hypoxic cages. Gene expression levels was assessed by quantitative real-time polymerase chain reaction. Protein expression was assessed by immunohistochemistry and enzyme-linked immunosorbent assay. Results: Ninety minutes after the shock induction, a vascular bed-specific, heterogeneous proinflammatory endothelial activation represented by E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1 expression was seen in kidney and lung. No differences in adhesion molecules between the spontaneously breathing and mechanically ventilated mice were found. Concentrations of the proinflammatory cytokines chemokine (C-X-C motif) ligand 1 (11.0-fold) and interleukin-6 (21.7-fold) were increased after 90 min of HS. Two hours of 6% oxygen did not induce the expression of E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1 in the kidneys and the lung. Conclusions: Hemorrhagic shock leads to an early and reversible proinflammatory endothelial activation in kidney and lung. HS-induced endothelial activation is not changed by mechanical ventilation during the shock phase. Hypoxia alone does not lead to endothelial activation. The observed proinflammatory endothelial activation is mostly ischemia- or reperfusion-dependent and not related to hypoxia.


American Journal of Physiology-renal Physiology | 2017

Corrigendum: Shock-induced stress induces loss of microvascular endothelial Tie2 in the kidney which is not associated with reduced glomerular barrier function (vol 297, pg F272, 2009)

M. van Meurs; Neng F. Kurniati; Francis M. Wulfert; Sigridur A. Ásgeirsdóttir; I. A. de Graaf; Simon C. Satchell; Peter W. Mathieson; Rianne M. Jongman; P. K. Kmpers; Jan G. Zijlstra; Peter Heeringa; Grietje Molema


F1000Research | 2012

Age dependent role of microvascular endothelial and polymorphonuclear cells in LPS induced renal failure

Francis M. Wulfert; M. van Meurs; Nf Kurniat; Rianne M. Jongman; Martin C. Houwertjes; Peter Heeringa; Jan G. Zijlstra; Mmrf Struys; Grietje Molema


Anesthesiology | 2011

Age Dependent Role of Microvascular Endothelial and Polymorphonuclear Cells in LPS Induced Renal Failure

Francis M. Wulfert; Matijs van Meurs; Michel Struys; Jan G. Zijlstra; Grietje Molema


Nederlands Tijdschrift voor Anesthesiologie | 2010

Increased renal microvascular endothelial responsiveness to LPS in aged mice

Francis M. Wulfert; M. van Meurs; Neng F. Kurniati; Rianne M. Jongman; Peter Heeringa; Jan G. Zijlstra; Mmrf Struys; Grietje Molema


Nederlands Tijdschrift voor Anesthesiologie | 2009

Renal microvascular endothelial activation in a mouse model of hemorrhagic shock: is the neutrophil a key player in organ dysfunction?

Francis M. Wulfert; M. van Meurs; Neng F. Kurniati; Mmrf Struys; Jan G. Zijlstra; Grietje Molema

Collaboration


Dive into the Francis M. Wulfert's collaboration.

Top Co-Authors

Avatar

Grietje Molema

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jan G. Zijlstra

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Matijs van Meurs

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neng F. Kurniati

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Peter Heeringa

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Rianne M. Jongman

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Leon Aarts

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ageeth J. Knol

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge