Network


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

Hotspot


Dive into the research topics where Joerg C. Kalff is active.

Publication


Featured researches published by Joerg C. Kalff.


Gastroenterology | 2014

Postoperative Ileus Involves Interleukin-1 Receptor Signaling in Enteric Glia

Burkhard Stoffels; Kristof J. Hupa; Susanne A. Snoek; Sjoerd H. van Bree; Kathy Stein; Timo Schwandt; Tim O. Vilz; Mariola Lysson; Cornelis van’t Veer; Markus P. Kummer; Veit Hornung; Joerg C. Kalff; Wouter J. de Jonge; Sven Wehner

BACKGROUND & AIMSnPostoperative ileus (POI) is a common consequence of abdominal surgery that increases the risk of postoperative complications and morbidity. We investigated the cellular mechanisms and immune responses involved in the pathogenesis of POI.nnnMETHODSnWe studied a mouse model of POI in which intestinal manipulation leads to inflammation of the muscularis externa and disrupts motility. We used C57BL/6 (control) mice as well as mice deficient in Toll-like receptors (TLRs) and cytokine signaling components (TLR-2(-/-), TLR-4(-/-), TLR-2/4(-/-), MyD88(-/-), MyD88/TLR adaptor molecule 1(-/-), interleukin-1 receptor [IL-1R1](-/-), and interleukin (IL)-18(-/-) mice). Bone marrow transplantation experiments were performed to determine which cytokine receptors and cell types are involved in the pathogenesis of POI.nnnRESULTSnDevelopment of POI did not require TLRs 2, 4, or 9 or MyD88/TLR adaptor molecule 2 but did require MyD88, indicating a role for IL-1R1. IL-1R1(-/-) mice did not develop POI; however, mice deficient in IL-18, which also signals via MyD88, developed POI. Mice given injections of an IL-1 receptor antagonist (anakinra) or antibodies to deplete IL-1α and IL-1β before intestinal manipulation were protected from POI. Induction of POI activated the inflammasome in muscularis externa tissues of C57BL6 mice, and IL-1α and IL-1β were released in ex vivo organ bath cultures. In bone marrow transplantation experiments, the development of POI required activation of IL-1 receptor in nonhematopoietic cells. IL-1R1 was expressed by enteric glial cells in the myenteric plexus layer, and cultured primary enteric glia cells expressed IL-6 and the chemokine monocyte chemotactic protein 1 in response to IL-1β stimulation. Immunohistochemical analysis of human small bowel tissue samples confirmed expression of IL-1R1 in the ganglia of the myenteric plexus.nnnCONCLUSIONSnIL-1 signaling, via IL-1R1 and MyD88, is required for development of POI after intestinal manipulation in mice. Agents that interfere with the IL-1 signaling pathway are likely to be effective in the treatment of POI.


Gastroenterology | 2009

Inhibition of p38 mitogen-activated protein kinase pathway as prophylaxis of postoperative ileus in mice.

Sven Wehner; Stefan Straesser; Tim O. Vilz; Dimitrios Pantelis; Thais Sielecki; Vidal F. de la Cruz; A. Hirner; Joerg C. Kalff

BACKGROUND & AIMSnPostoperative ileus, an iatrogenic complication of abdominal surgery, is mediated by severe inflammation of the tunica muscularis. Macrophages that reside in the muscularis have important roles in initiating the inflammation. We investigated whether activation of the p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase is involved in the genesis of postoperative ileus, and whether p38-MAPK inhibition by the macrophage-specific inhibitor semapimod prevents intestinal dysmotility.nnnMETHODSnPostoperative ileus was induced by intestinal manipulation of the small bowel in mice. Protein kinase phosphorylation was assessed by immunoblotting of muscularis externa preparations. Proinflammatory gene expression was quantified by real-time polymerase chain reaction. Myeloperoxidase histochemistry for neutrophils was performed in jejunal segments. Nitric oxide production was measured by Griess reaction in smooth-muscle organ culture supernatants. Jejunal contractility was assessed within an organ bath setup. Intestinal motility was analyzed by gastrointestinal and colonic transit measurements.nnnRESULTSnHigh levels of p38-MAPK and stress-activated protein kinase phosphorylation were observed immediately after intestinal manipulation. Semapimod treatment led to a significant decrease of p38-MAPK phosphorylation in macrophages; proinflammatory gene expression of macrophage inflammatory protein-1alpha, interleukin-6, monocyte chemoattractant protein-1, and intercellular adhesion molecule-1; and neutrophil infiltration. Furthermore, semapimod completely abrogated nitric oxide production within the tunica muscularis. Subsequently, semapimod prevented the suppression of smooth muscle contractility and small intestinal and colonic motility after intestinal manipulation.nnnCONCLUSIONnA single preoperative semapimod administration prevents intestinal macrophage activation and subsequent gastrointestinal dysmotility induced by abdominal surgery. Semapimod inhibits p38-MAPK and nitric oxide production in macrophages, making it a promising strategy for prophylaxis of postoperative ileus.


Langenbeck's Archives of Surgery | 2012

Immune mediators of postoperative ileus

Sven Wehner; Tim O. Vilz; Burkhard Stoffels; Joerg C. Kalff

Clinical backgroundIn all patients undergoing abdominal surgery, a transient phase of interruption of bowel motility, named postoperative ileus (POI) occurs. POI is often accepted as an unavoidable “physiological” response and a self-limiting complication after surgery although it has a significant impact on patient morbidity with prolonged hospitalization and increased costs. Annual economic burden has been estimated as much as US


Langenbeck's Archives of Surgery | 2010

The effect of sealing with a fixed combination of collagen matrix-bound coagulation factors on the healing of colonic anastomoses in experimental high-risk mice models

Dimitrios Pantelis; Anke Beissel; Philip Kahl; Sven Wehner; Tim O. Vilz; Joerg C. Kalff

1.47 billion in the USA (Iyer et al. in J Manag Care Pharm 15(6):485–494, 2009).PathophysiologyThe pathophysiology has been elucidated within the last decades, demonstrating that both, neurogenic and inflammatory mechanisms are involved in response to the surgical trauma. It is now generally accepted that POI pathogenesis processes in two phases: a first neurogenic phase is accountable for the immediate postoperative impairment of bowel motility. This is followed by a second immunological phase that can last for days and mainly affects strength and length of POI. More recent findings demonstrate a bidirectional interaction between the nervous and the immune system, and this interaction significantly contributed to our present understanding of POI pathophysiology. Although nerval mechanisms have a significant impact in the early phase of POI, the contribution of the immune system and subsequently its manipulation has risen as the most promising strategy in prevention or treatment of the clinically relevant prolonged form of POI.AimsThe present manuscript will give an update on the inflammatory responses, the involved cell types, and participating immune mediators in POI.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Mechanical strain and TLR4 synergistically induce cell-specific inflammatory gene expression in intestinal smooth muscle cells and peritoneal macrophages

Sven Wehner; Bettina M. Buchholz; Silke Schuchtrup; Anatol Rocke; Nico Schaefer; Mariola Lysson; A. Hirner; Joerg C. Kalff

PurposeExperimental and clinical studies on the sealing of colorectal anastomoses in order to reduce the rate of leakage have previously been performed with divergent results. However, comparatively few studies have been performed on anastomotic healing using a fibrin glue-coated patch. The aim of this experimental basic scientific study in mice was to investigate the effect of fibrin glue-coated collagen patches on the healing process of colonic anastomoses in situations of adverse healing process (technical deficiency and peritonitis).MethodsColonic anastomoses were carried out in 206 mice and randomized into six groups (I: complete anastomoses, II: sealed complete anastomoses, III: incomplete anastomoses, IV: sealed incomplete anastomoses, V: complete anastomoses in the presence of bacterial peritonitis, VI: sealed complete anastomoses in the presence of bacterial peritonitis). Tissues from the anastomoses were removed and used for functional, histochemical, molecular, and biochemical investigations.ResultsThe evaluation of postoperative course data revealed the beneficial effect of additional sealing with a fixed combination of collagen matrix-bound coagulation factors I and IIa (Tachosil®, Nycomed Austria, Linz) in high-risk experimental anastomotic healing. Sealing incomplete anastomoses resulted in significantly lower lethality and leakage rates, as well as significantly higher bursting pressure values and histopathologic scores. Collagen 1 and 3 expressions and hydroxyproline concentrations are greatly increased with additional sealing in all high-risk anastomoses.ConclusionsIn our current model, we demonstrate that additionally sealing high-risk experimental colonic anastomoses provides a positive effect on the healing process. The effect on the molecular level in particular seems to be essential and requires further experimental studies to evaluate the mechanism.


American Journal of Surgery | 2010

Perioperative management and outcome of general and abdominal surgery in hemophiliacs.

G. Goldmann; Yaroslava Holoborodska; Johannes Oldenburg; Nico Schaefer; Tobias Hoeller; Jens Standop; Joerg C. Kalff; A. Hirner; Marcus Overhaus

Mechanical trauma of the gut is an unavoidable event in abdominal surgery. Former studies demonstrated that intestinal manipulation induces a strong inflammation within the tunica muscularis. We hypothesized that mechanical strain initiates or aggravates proinflammatory responses in intestinal smooth muscle cells (iSMC) or macrophages. First, an appropriate isolation and culture method for neonatal rat iSMC was established. Purified iSMC and primary peritoneal macrophages (pMacs) were subjected to static or cyclic strain, and gene expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), IL-6, and IL-1β was analyzed by quantitative PCR. Supernatants from stretched iSMC were transferred to untreated pMacs or contrariwise, and medium transfer-triggered inflammatory gene expression was measured in unstretched cells. Finally, we investigated the synergistic effect of static strain on LPS-induced proinflammatory gene expression. Although cyclic strain failed, static strain significantly induced iNOS, COX-2, and IL-1β mRNA in iSMC. pMacs showed an increase in all inflammatory genes investigated as well as macrophage inflammatory protein (MIP)-1α and MIP-2 mRNA after static strain. Both cell entities liberated unknown mediators in response to stretch that mutually stimulated iNOS gene expression. Finally, mechanostimulation amplified LPS-induced iNOS and IL-1β gene expression in iSMC as well as COX-2 and IL-6 mRNA in pMacs. In conclusion, static strain initiates proinflammatory gene expression in iSMC and pMacs and triggers a bidirectional paracrine communication between both cultured cell entities via the liberation of unknown mediators. Furthermore, static strain synergistically operates with Toll-like receptor 4 ligation in a cell-specific manner. Hence, this study demonstrates that mechanical strain functions as an immunomodulatory stimulus in abdominal cells.


Langenbeck's Archives of Surgery | 2012

Preoperative short-term parenteral administration of polyunsaturated fatty acids ameliorates intestinal inflammation and postoperative ileus in rodents

Sven Wehner; Katharina P. Meder; Tim O. Vilz; Birgit Alteheld; Peter Stehle; Thomas Pech; Joerg C. Kalff

BACKGROUNDnThe aim of the current study was to investigate perioperative management and outcome of surgery in hemophiliacs.nnnMETHODSnFifty-five hemophiliacs underwent surgery (appendectomy, cholecystectomy, inguinal hernia repair, hemorrhoidectomy). Surgical procedures in hemophiliacs and matched pairs were analyzed for duration of surgery, drainages, hospital stay, factor use (VIII, IX), and complications. Factor substitution was analyzed. Mann-Whitney U and Kruskal-Wallis tests were used (P < .05).nnnRESULTSnNo significant differences were found for duration of drains and operation time in hemophiliacs versus matched pairs. Significance for duration of hospital stay compared with controls was found in hemophiliacs for appendectomy, inguinal hernia repair, and hemorrhoidectomy but not for cholecystectomy. In both groups, complications were low without significant differences.nnnCONCLUSIONSnThis study found no significant differences in perioperative data and postoperative outcome in hemophiliacs compared with nonhemophiliacs due to the excellent perioperative interdisciplinary management at our Hemophilia Center with prolonged hospital stay in hemophiliacs.


Transplantation | 2012

Combination therapy of tacrolimus and infliximab reduces inflammatory response and dysmotility in experimental small bowel transplantation in rats.

Thomas Pech; Jun Fujishiro; Tobias Finger; Ichiro Ohsawa; Michael Praktiknjo; Kareem Abu-Elmagd; Martin von Websky; Marcus Overhaus; Joerg C. Kalff; Nico Schaefer

PurposeAbdominal surgery results in an inflammation of the intestinal muscularis externa (ME), subsequently leading to postoperative ileus (POI). Polyunsaturated fatty acids (PUFA) are known to modulate inflammation. The aim of this study was to analyze the effect of preoperative parenteral administration of marine (n-3) or soybean (n-6) PUFA lipid emulsions (PUFA-LE) on POI and tissue fatty acid profiles.MethodsRodents underwent intestinal manipulation (IM) after 5xa0days of parenteral administration of 10-mL/kg body weight saline, (n-3), or (n-6) PUFA-LE. Sham animals received saline treatment without IM. In rats, postoperative inflammation was quantified by ME neutrophil levels and NO production in organ culture, and ME function was determined by an in vitro contractility measurement. Additionally, in vivo gastrointestinal transit (GIT) was analyzed in mice. Lipopolysaccharide-induced IL-6 expression of rat bone marrow-derived mononuclear cells and ME was analyzed. Fatty acids were measured by gas chromatography in rat blood, bone marrow cells, and ME.ResultsThe (n-3) PUFA-LE reduced neutrophil levels and NO production after IM and improved in vitro jejunal contractility and GIT time. The (n-6) PUFA-LE significantly reduced postoperative inflammation and tended to improve intestinal motility (Pu2009<u20090.06). Interestingly, (n-6) PUFA-LE significantly reduced the levels of arachidonic acid in ME (−63%), while (n-3) PUFA-LE reduced arachidonic acid (−20%) and additionally raised EPA (+550%).ConclusionShort-term preoperative parenteral administration of (n-3) or (n-6) PUFA-LE significantly alters tissue-specific fatty acid profiles. Preoperative parenteral PUFA-LE supplementation, preferably by marine (n-3) PUFA, ameliorates postoperative intestinal inflammation and dysmotility and could be a promising therapeutic option in POI prophylaxis.


Journal of Visualized Experiments | 2012

Functional assessment of intestinal motility and gut wall inflammation in rodents: analyses in a standardized model of intestinal manipulation.

Tim O. Vilz; Marcus Overhaus; Burkhard Stoffels; Martin von Websky; Joerg C. Kalff; Sven Wehner

Background. Intestinal transplantation initiates a functionally relevant inflammatory response by activation of resident macrophages within the muscularis associated with dysmotility. Infliximab is used successfully as a potent anti-inflammatory agent for the treatment of chronic inflammatory bowel diseases and as rescue therapy in acute steroid-resistant rejection in selected settings in clinical small bowel transplantation. We hypothesize that additional perioperative treatment with infliximab diminishes initiation of the inflammatory cascade and improves motility in small bowel grafts using a standard tacrolimus immunosuppressive protocol. Methods. Orthotopic intestinal transplantation was performed in rats. In two treatment groups (24/168 hr), infliximab was administered intravenously directly after reperfusion and tacrolimus was injected intramuscularly after transplantation and once a day. Two other treatment groups (24/168 hr) received standard immunosuppressive therapy with tacrolimus. Isogenic and allogenic transplanted vehicle-treated animals (24/168 hr) and native gut served as control. Results. Infliximab-treated grafts exhibited significantly less leukocyte infiltration at 24/168 hr after transplantation and at 168 hr significantly less apoptosis in the tunica muscularis compared with tacrolimus monotherapy. Additional infliximab treatment resulted in increased smooth muscle contractility (30%) after 24 hr compared with tacrolimus control. Conclusions. Dysmotility of transplanted small bowel results from reperfusion injury and acute rejection. Additional perioperative treatment with infliximab reduces early unspecific inflammatory responses and complements immunosuppressive therapy with tacrolimus.


Surgery | 2010

Transient perioperative pharmacologic inhibition of muscularis macrophages as a target for prophylaxis of postoperative ileus does not affect anastomotic healing in mice.

Dimitrios Pantelis; Mustapha Sundifu Kabba; Jutta Kirfel; Philip Kahl; Sven Wehner; Reinhard Buettner; A. Hirner; Joerg C. Kalff

Inflammation of the gastrointestinal tract is a common reason for a variety of human diseases. Animal research models are critical in investigating the complex cellular and molecular of intestinal pathology. Although the tunica mucosa is often the organ of interest in many inflammatory diseases, recent works demonstrated that the muscularis externa (ME) is also a highly immunocompetent organ that harbours a dense network of resident immunocytes.(1,2) These works were performed within the standardized model of intestinal manipulation (IM) that leads to inflammation of the bowel wall, mainly limited to the ME. Clinically this inflammation leads to prolonged intestinal dysmotility, known as postoperative ileus (POI) which is a frequent and unavoidable complication after abdominal surgery.(3) The inflammation is characterized by liberation of proinflammatory mediators such as IL-6(4) or IL-1β or inhibitory neurotransmitters like nitric oxide (NO).(5) Subsequently, tremendous numbers of immunocytes extravasate into the ME, dominated by polymorphonuclear neutrophils (PMN) and monocytes and finally maintain POI.(2) Lasting for days, this intestinal paralysis leads to an increased risk of aspiration, bacterial translocation and infectious complications up to sepsis and multi organ failure and causes a high economic burden.(6) In this manuscript we demonstrate the standardized model of IM and in vivo assessment of gastrointestinal transit (GIT) and colonic transit. Furthermore we demonstrate a method for separation of the ME from the tunica mucosa followed by immunological analysis, which is crucial to distinguish between the inflammatory responses in these both highly immunoactive bowel wall compartments. All analyses are easily transferable to any other research models, affecting gastrointestinal function.

Collaboration


Dive into the Joerg C. Kalff'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge