Skip to main content
Erschienen in: Journal of Gastrointestinal Surgery 5/2007

Open Access 01.05.2007

The Use of Animal Models to Study Bacterial Translocation During Acute Pancreatitis

verfasst von: L. P. van Minnen, M. Blom, H. M. Timmerman, M. R. Visser, H. G. Gooszen, L. M. A. Akkermans

Erschienen in: Journal of Gastrointestinal Surgery | Ausgabe 5/2007

Abstract

Infection of pancreatic necrosis with intestinal flora is accepted to be a main predictor of outcome during severe acute pancreatitis. Bacterial translocation is the process whereby luminal bacteria migrate to extraintestinal sites. Animal models were proven indispensable in detecting three major aspects of bacterial translocation: small bowel bacterial overgrowth, mucosal barrier failure, and disturbed immune responses. Despite the progress made in the knowledge of bacterial translocation, the exact mechanism, origin and route of bacteria, and the optimal prophylactic and treatment strategies remain unclear. Methodological restrictions of animal models are likely to be the cause of this uncertainty. A literature review of animal models used to study bacterial translocation during acute pancreatitis demonstrates that many experimental techniques per se interfere with intestinal flora, mucosal barrier function, or immune response. Interference with these major aspects of bacterial translocation complicates interpretation of study results. This paper addresses these and other issues of animal models most frequently used to study bacterial translocation during acute pancreatitis.

Introduction

Experimental models of acute pancreatitis exist for almost 150 years, with Claude Bernard first describing experimental pancreatitis by injection of bile and olive oil into the pancreatic duct of a rabbit.1 Ever since, animal experiments were indispensable in providing insight in pathophysiology and treatment of acute pancreatitis. Experimental studies have major advantages over clinical studies, such as the availability of study subjects, standardization of disease severity, ability to perform invasive tests, extensive tissue sampling, and the possibility to test prophylactic treatment strategies.2 Despite these advantages, some major aspects of the pathophysiology of acute pancreatitis remain unclear, mortality in severe acute pancreatitis is still as high as 5–28%, and optimal treatment strategies remain a topic of debate.3,4
In 1986, Beger et al. demonstrated a link between the intestinal flora, infection of pancreatic necrosis, and clinical outcome in patients with severe acute pancreatitis.5 At the present time, infection of pancreatic necrosis is still regarded to be a main predictor of outcome during severe acute pancreatitis, and bacterial translocation of intestinal flora is considered to be the cause.4
Changes in intestinal motility and the associated shift of intestinal flora, mucosal barrier function, and the immune system were identified as pivotal aspects of bacterial translocation during acute pancreatitis.611 This has greatly increased the understanding of bacterial translocation, but better insight into the exact mechanism of bacterial translocation and subsequent infection of pancreatic necrosis is needed to develop adequate prophylaxis and treatment strategies for patients with severe acute pancreatitis.
A multitude of animal models were used to study the mechanism of bacterial translocation, including radiolabeling, plasmid-labeled bacteria, or fluorescent beads.1215 Despite all these efforts, however, the exact origin, route, and mechanism of bacterial translocation causing infection of pancreatic necrosis are still unclear. The main reason for this uncertainty is the lack of an “ideal” animal model of acute pancreatitis to study pathophysiology of bacterial translocation and its treatment. The ideal model should be minimally invasive, standardized, reproducible, and resemble etiology, pathophysiology, disease course, and outcome of clinical acute pancreatitis, including response to treatment.2 Experimental models used to study bacterial translocation in acute pancreatitis and its treatment all seem to have methodological restrictions that complicate the interpretation of study results. In 2000, Foitzik et al. reviewed the use of animal models of acute pancreatitis and their suitability for evaluating therapy and concluded that animal models should be designed to mimic etiology and clinical course of human pancreatitis to increase their value.2 In addition, we would like to discuss the value animals studies and experimental models of acute pancreatitis have in face of their interference with one or more of the known aspects of bacterial translocation: intestinal motility and flora, mucosal barrier function, or the immune system.
The aim of this paper is to provide useful insights into the use of animal models to study bacterial translocation during acute pancreatitis, in the light of current knowledge of pathophysiology.

Animal Species and Housing Conditions

Before the late 1970s, larger laboratory animals such as dogs were predominantly used to study acute pancreatitis. But since the introduction of models of acute pancreatitis in small laboratory animals, mice or rats are generally favored for financial and ethical or practical reasons. Because of physiological and anatomical differences between species, choice of laboratory animal has important implications on the study results and extrapolation to the human situation.
Intestinal flora differs between animal species, largely depending on dietary demands and anatomical differences of the gastrointestinal tract and habits.1618 The protein-rich diet of dogs or cats results in lower counts of endogenous lactobacilli and higher counts of potential pathogens (e.g., clostridia species), compared to rats or mice with fiber-rich diets. Coprophagy, demonstrated by most rodents, also influences intestinal flora, resulting in higher counts of gram-negative bacteria in the proximal gastrointestinal tract.19,20 Also, rats and mice are often bred and kept under specific pathogen-free conditions, introducing modifications of intestinal flora.
Intestinal barrier function also differs between species. In an experiment comparing small intestinal permeability between humans and rats, significant interspecies variation in urinary recovery of orally delivered mannitol was observed.21
Anatomical differences between species should also be considered. The relative size of the jejunum, ileum, cecum, and colon of different laboratory animals can influence origin and route of bacterial translocation during acute pancreatitis. In humans, retroperitoneal connections between the intestines and pancreas can greatly affect the clinical course of the disease.22 Similar to humans, the dog pancreas is situated retroperitoneally. Rat and mouse pancreata, however, are almost fully enveloped by peritoneum, resembling a more intraperitoneal localization. Variation in retroperitoneal connections between intestines and the pancreas offers different routes for bacteria to translocate without being exposed to intraperitoneal immune cells.23
Experiments using small animals (e.g., mouse or rat) usually incorporate a larger number of animals compared to experiments with large laboratory animals (e.g., cat or dog). The use of a larger number of small laboratory animals improves statistical power of an experiment. On the other hand, the use of larger animals could resemble human pathophysiology better, but a smaller number of animals means lower statistical power and increased potential false negative or false positive results.

Models of Acute Pancreatitis

An abundance of animal models of acute pancreatitis is used to investigate bacterial translocation. Only models most frequently used for this purpose will be discussed. Baseline characteristics of the discussed models and their potential effects on intestinal flora, mucosal barrier, and immune function are summarized in Tables 1 and 2.
Table 1
Characteristics of Several Animal Models of Acute Pancreatitis
Model
Animal Species
Pancreatic Necrosis
Pancreatic Infection
Mortality
Invasiveness
Duodenal loop24,25
Rat
No
Considerable
High
Laparotomy
Choline-deficient diet3032
Mouse
Yes
Little
High
Minimal
Duct ligation3437
Rat/opossum
No/Yes
Little
Low
Laparotomy
Cerulein44
Mouse/rat
Yes/No
Little
Low
Minimal
Duct perfusion48
Rat/dog/pig
Yes
Considerable
Moderate to high
Laparotomy
Duct perfusion + cerulein52
Rat
Yes
Considerable
Moderate
Laparotomy
Table 2
Aspects of Bacterial Translocation and Potential Confounding Factors of Animal Models
Aspect
Confounding Factor
Model
Intestinal motility and flora
Animal species
Potentially all models
Housing conditions (SPF)
Potentially all models
Diet
CDE diet
Analgesics
Invasive models
Laparotomy
Invasive models
Bile flow
Duct ligation
Cerulein
Cerulein models
Intestinal manipulation
Invasive models
Mucosal barrier function
Stress
Potentially all models
Diet
CDE diet
Anesthetics
Invasive models
Pancreatic proteases
Duct ligation
Intestinal manipulation/puncture
Duct perfusion
Immune system
Stress
Potentially all models
Diet
CDE diet
Disease course/severity
Species-dependent
Obstructive jaundice
Duct ligation, duodenal loop
Intestinal manipulation
Invasive models

Duodenal Loop

Closing the duodenal lumen proximally and distally to the papilla of Vater results in reflux of the duodenal contents enclosed in the loop, including bile and pancreatic secretions, into the biliopancreatic duct.24 In rats, this leads to acute pancreatitis of varying severity.25 Discontinuation of the gastrointestinal tract leads to mucosal atrophy and functional changes to the mucosal barrier.26 Furthermore, obstruction of bile flow into the intestine was shown to reduce intestinal motility, causing small bowel bacterial overgrowth and increased bacterial translocation.2729 Another major downside is the occurrence of reflux of duodenal contents, including bacteria, into the biliopancreatic duct. These obvious drawbacks of this model in experiments concerning bacterial translocation are the cause of its limited popularity.

Ethionine-supplemented Choline Deficiency

Lombardi et al.30 described severe acute pancreatitis in young female mice after feeding a choline-deficient, ethionine-supplemented (CDE) diet.31 Acute hemorrhagic pancreatitis ensues, as well as diffuse intraperitoneal fat necrosis and several systemic effects such as acidosis, hypoxia, and hypovolemia. In this model, mortality ranges from 0 to 100% after 4 days and can be controlled by varying the duration of the choline-deficient diet.32 To ensure homogeneity and reproducibility, sex, age, and weight of the mice have to be closely matched, as well as food intake of all animals.32
Apart from these practical downsides of the model, systemic complications unrelated to pancreatitis (e.g., parotitis and fatty liver disease) render the model less useful for investigating systemic events (e.g., immune response) of acute pancreatitis.31 Little is known of the effect of ethionine suppletion or choline deficiency on intestinal flora or mucosal barrier function. But the most important drawback of this model to study bacterial translocation is the low incidence of pancreatic infection (3–8%), even in severe necrotizing pancreatitis.33

Biliopancreatic Duct Ligation

In the duct ligation model, the common biliopancreatic duct is surgically clipped or tied at the sphincter of Oddi complex. The resulting obstruction of pancreatic secretions and potential biliary reflux into the pancreatic duct produce moderate pancreatitis, characterized by edema, moderate inflammation and hemorrhage, fat necrosis, and minimal acinar cell necrosis. Only in the American opossum does biliopancreatic duct ligation leads to severe acute pancreatitis with considerable necrosis.3437
This model of acute pancreatitis greatly interferes with the pathophysiology of bacterial translocation. Obstruction of bile flow into the intestine causes small bowel bacterial overgrowth and bacterial translocation.28 Also, exclusion of pancreatic proteases in the gut lumen alters intestinal permeability.38,39 Apart from effects on the intestinal flora and mucosal barrier function, obstruction-induced jaundice also causes impairment of the immunesystem.4042 These effects complicate the interpretation of bacteriological results to study bacterial translocation.

Cerulein Infusion

Infusion of low doses of cerulein, a cholecystokinin analog, enhances production of pancreatic exocrine cell secretions without cell necrosis. In most species, infusion of supramaximal doses results in a decrease of secretion and acute pancreatitis with interstitial edema and inflammatory cell infiltration.43 In mice, cerulein causes severe acute pancreatitis with necrosis of 40% of acinar cells.44 In rats and other animals, however, cerulein-induced pancreatitis is usually mild and generally self-limiting. Moreover, pigs are reported to be insensitive to cerulein hyperstimulation.45 It should be noted that cerulein is known to affect intestinal motility. Studies investigating the use of cerulein in man have shown absence of recognizable migrating motor complexes with decreased colonic transit time.46 In general, experimental acute pancreatitis is associated with reduced small bowel motility, resulting in small bowel bacterial overgrowth and increased bacterial translocation to extraintestinal sites.6,47 Thus, cerulein may interfere with intestinal flora by altering intestinal motility. Investigators should keep this in mind when designing a study and interpreting study results.

Biliopancreatic Duct Perfusion

Duct perfusion models are currently the most popular models of acute pancreatitis. Induction of acute pancreatitis involves infusion of bile, bile salts with or without bacteria, or activated pancreatic enzymes into the (bilio-)pancreatic duct. Early experiments mainly involved dogs, but currently, rats are used most frequently. Severity and reproducibility of acute pancreatitis and ensuing bacteriological results strongly depend on infusate, infusion pressure, volume, and time.48
The most commonly used infusates are solutions containing various concentrations of bile salts of varying hydrophobicity. Both chemical and pressure effects of infusion were suggested to play a major role in the pathogenesis of pancreatitis in perfusion models.48,49 In both chemical- and pressure-induced pancreatitis, destruction of the pancreatic duct mucosal barrier is the key event. This is followed by pancreatic edema, autolysis, reduction of pancreatic blood flow, and, in severe cases, destruction of pancreatic parenchyma and formation of pancreatic necrosis.50 Uncontrolled pressure-related damage causes variation in severity of the induced acute pancreatitis between study subjects, and thus should be avoided. Several experiments were performed to assess maximal pancreatic duct pressure before rupture of the duct epithelium causing increased and uncontrolled severity of acute pancreatitis. Data are conflicting, with rupture pressures varying from 15 to 82 mmHg.48,49,51,52 A maximum infusion pressure of 30 to 50 mmHg is currently accepted for rat models.
Perfusion is usually performed by puncturing the duodenum and cannulating the papilla of Vater. The introduction of duodenal bacteria, through the papilla of Vater into the biliopancreatic duct could potentially be a confounding factor in transduodenal duct perfusion models. It was demonstrated, however, that significant bacterial infection of the pancreas (>1 × 102 colony forming units per gram) because of the surgical procedure does not occur.53
Advantages of this model are the quick procedure of acute pancreatitis induction and the reproducibility of results. Other than duodenal puncturing and intestinal handling during surgery, both potentially affecting mucosal barrier function, no direct effects on intestinal flora or immune function are expected in this model.

Biliopancreatic Duct Injection and Cerulein Hyperstimulation

The combination of retrograde infusion of bile salts with superimposed cerulein hyperstimulation in rats was introduced by Schmidt et al. and was advocated as “a better model for evaluating therapy.”52 Although the disadvantages described for biliopancreatic duct injection and cerulein hyperstimulation all apply to this model, it was proven a very valuable model to examine bacterial translocation and treatment strategies. The major advantages are that histological and qualitative bacteriological results as well as reaction to treatment and disease course resemble human acute pancreatitis more closely than other models.2,52 Although proven a very valuable model, potential model-related confounding factors as described above should always be kept in mind when interpreting results.

Disease Course

Especially in the severe form of acute pancreatitis, systemic events can be divided into two phases: early proinflammatory and late immunosuppressive.54 In severe acute pancreatitis, the early phase is associated with a systemic inflammatory response syndrome (SIRS), potentially leading to multiple organ failure and early mortality. The late phase is characterized by immunosuppression, providing opportunity for infectious complications (e.g., infection of pancreatic necrosis) associated with sepsis and late mortality.2,55 Laboratory animal species and experimental models, however, each show their own disease course of acute pancreatitis.
Animal models were mainly used to investigate the early phase of acute pancreatitis.56 However, the model described by Schmidt et al. seems the most appropriate to investigate early and late systemic complications, considering that both phases can be discerned.52,57 In this model, infection of pancreatic necrosis progresses at least until 96 h. When taking into account that disease course is more rapid in small rodents, timing could well correlate with data on the course of severe acute pancreatitis in humans, as described by Foitzik et al.2, Beger et al.,4 and Lankisch et al.58

Severity

Pancreatic necrosis is produced in several animal models of acute pancreatitis (Table 1). On the other hand, only duct perfusion, with or without superimposed cerulein hyperstimulation, and murine CDE models demonstrate mortality comparable to human necrotizing acute pancreatitis.32,52,59 Models with high early mortality may be useful to investigate early phase systemic inflammatory response and organ failure, but are less adequate to investigate late infectious complications and associated (multiple) organ failure.
In most models, necrosis needs to be present for pancreatic infection to occur. It needs to be noted that this does not apply for the duodenal loop model in which reflux of duodenal contents into the biliopancreatic duct occurs.60 In contrast, the murine CDE model produces elaborate necrosis, but is associated with very low rates of pancreatic infection.33

Culturing, Controls, and Route of Bacterial Translocation

In all animal models, factors such as analgesia, anesthesia, or surgical techniques can influence bacteriological results. Morphine-like analgesics have a significant effect on bowel motility and cause bacterial overgrowth and translocation to extraintestinal sites.61 The anesthetic pentobarbital was suspected to be a factor in promoting bacterial translocation in a model of hemorrhagic shock.62
Also, stress causes mucosal barrier failure and bacterial translocation.63 Surgical procedures are stressful events, but animal transport or handling alone could potentially cause stress-induced bacterial translocation. The influence of stress on adrenaline and corticosteroid levels could have its own effect on the function of the immune system, potentially influencing the systemic reaction to acute pancreatitis and bacterial translocation.
Proper sterile surgical techniques are very important when investigating bacterial translocation. If abdominal surgery is involved, control cultures of the peritoneal cavity to trace surgical contamination are of special importance. If peritoneal cultures are found to be positive, extra caution should be taken with interpretation of bacteriological analysis of abdominal organs. In case of surgical contamination or transperitoneal bacterial translocation, the peritoneal covering of the organ samples might be the cause of positive organ cultures, not the bacterial colonization in the organ itself (false positive culture).
Puncturing the duodenum in duct infusion models hypothetically causes spillage of duodenal contents onto the peritoneum, covering all abdominal organs. In rats, however, duodenal contents usually have low bacterial counts, mainly consisting of nonpathogenic lactobacilli only. On the other hand, a duct infusion study by Cicalese et al. reported positive peritoneal cultures at time of induction of pancreatitis of 16.6 to 33.3% of the studied rats.15 Literature review of different animal models fairly frequently shows positive peritoneal cultures at the time of termination and organ sample collection of rats with acute pancreatitis. Positive peritoneal cultures are observed varying from 0–10% in minimally invasive models of acute pancreatitis (cerulein injection, CDE diet) to 8–100% in more invasive models (duct perfusion with or without cerulein hyperstimulation).6,14,15,6466

Discussion

Changes in intestinal motility and flora, mucosal barrier function, and immune response were established as pivotal aspects in the process of bacterial translocation during acute pancreatitis. Early after the onset of acute pancreatitis, neurohormonal effects result in reduced small bowel motility.6 This causes stasis of luminal contents and small bowel bacterial overgrowth with potential pathogens, including Escherichia coli and Enterococcus species. The abundant presence of luminal pathogens forms a challenge for the mucosal barrier. Furthermore, pancreatitis-associated reduced intestinal blood flow results in mucosal ischemia and reperfusion damage.6769 Luminal bacteria, normally held at bay by the mucosal barrier, now have opportunity to penetrate into the intestinal epithelium. Local intestinal inflammation follows, further compromising mucosal barrier function. Pancreatitis and ensuing intestinal inflammation both contribute to a systemic proinflammatory response (SIRS), with damaging effects on distant organs.70,71 If the systemic response is severe, multiple organ dysfunction syndrome (MODS) might follow.72,73 If the patient survives the early phase, counterregulatory immunological pathways releasing anti-inflammatory cytokines result in a refractory state characterized by immunosuppression.74,75 Persistent immunosuppression will render the patient liable for infection of pancreatic necrosis. Multiple organ dysfunction syndrome caused by infectious complications is considered accountable for so-called late mortality or “late septic death.”74,76
Although animal models were proven indispensable in acute pancreatitis research, model-related problems are most likely the reason for important questions on pathophysiology and treatment strategies to remain unanswered. Current topics of debate include the route and origin of bacterial translocation and optimal prophylaxis and treatment strategies.
Several different routes of bacterial translocation were described and have directed efforts for many prophylactic and therapeutic strategies. Webster et al. showed bacteremia to occur early after induction of acute pancreatitis in CDE-induced acute pancreatitis, suggesting a hematogenous route.77 Likewise, rapid passage of bacteria into the blood was found in other models of acute pancreatitis.78 On the other hand, Runkel et al. found bacteria migrating to lymph nodes before their translocation to distant sites in a duct ligation model, suggesting a lymphogenous route.79 Widdison et al. suggested transperitoneal translocation of bacteria originating from the colon in a feline model of severe necrotizing pancreatitis.80 Other study groups, including our own, have provided proof of the role of the small bowel in the pathophysiology of bacterial translocation in acute pancreatitis or after morphine administration.6,61,81
The model of duct perfusion and cerulein hyperstimulation described by Schmidt et al. was proven very useful because it resembles human disease quite well, considering its biphasic disease course, pancreatic histology, “moderate” mortality, and the bacterial spectrum in pancreatic necrosis.52 However, whether a confounder is introduced by puncturing the duodenum and cannulating the biliopancreatic duct is unknown. Therefore, to ensure quality of the presented study results, control cultures of the peritoneal cavity should be done when organ samples are analyzed bacteriologically. Peritoneal bacteria can potentially affect bacteriological analysis of all abdominal tissues. Widdison et al. washed abdominal samples before analysis, but this is not commonly performed.80 A pilot study by Arendt et al. showed that washing removed 94–97% of intraperitoneally injected bacteria.23 Immunohistologically localizing bacteria can help clarify if positive cultures of abdominal tissues are because of peritoneally located bacteria or actual bacterial colonization in the underlying organ tissue.
When experimentally evaluating therapy, treatment often starts before induction of acute pancreatitis. Obviously, this is an important reason why results cannot directly be translated to the clinical situation. On the other hand, these experimental studies provide proof of principle concerning the tested therapy. If prophylactically successful, the tested treatment strategy might be beneficial when started after the onset of acute pancreatitis and should therefore be further investigated. On the other hand, the faster course of acute pancreatitis in rodent models provides only a very short treatment window between the onset of the disease and early or late phase complications. This may lead to false negative effects of the therapy tested.
In conclusion, animal models of acute pancreatitis are indispensable tools, but model-related drawbacks often interfere with one or more pathophysiological aspects of bacterial translocation, complicating interpretation of results. When the ideal model of acute pancreatitis is not at hand, it is of major value that numerous alternatives are available. But with each experimental hypothesis, special care should be taken to select the most suitable model. Despite all the experimental work done, the route by which pancreatic infection occurs and gives rise to septic complications and mortality has not yet fully been elucidated. Optimal prophylactic and treatment strategies are also still widely debated. In the future, animal models will undoubtedly provide increasing understanding of these subjects, but model-related drawbacks should always be kept in mind when designing a study or when interpreting results.

Acknowledgments

L.P. van Minnen received financial support from Astra Zeneca, Research and Development, Mölndal, Sweden. H.M. Timmerman received funding from Winclove Bio Industries B.V., Amsterdam, The Netherlands. Part of this study was supported by Senter, an agency of the Dutch Ministry of Economic Affairs (grant no. TSGE3109). Supporting institutions were not involved in design, performance, or publication of this study.
Open Access This is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License ( https://​creativecommons.​org/​licenses/​by-nc/​2.​0 ), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Bernard C. Leçons de Physiologie Experimentale 2 (viii). Paris: Baillèire, 1856, pp 504–512. Bernard C. Leçons de Physiologie Experimentale 2 (viii). Paris: Baillèire, 1856, pp 504–512.
2.
Zurück zum Zitat Foitzik T, Hotz HG, Eibl G, Buhr HJ. Experimental models of acute pancreatitis: are they suitable for evaluating therapy? Int J Colorectal Dis 2000;15(3):127–135.PubMedCrossRef Foitzik T, Hotz HG, Eibl G, Buhr HJ. Experimental models of acute pancreatitis: are they suitable for evaluating therapy? Int J Colorectal Dis 2000;15(3):127–135.PubMedCrossRef
3.
Zurück zum Zitat Beger HG, Rau B, Isenmann R. Natural history of necrotizing pancreatitis. Pancreatology 2003;3(2):93–101.PubMedCrossRef Beger HG, Rau B, Isenmann R. Natural history of necrotizing pancreatitis. Pancreatology 2003;3(2):93–101.PubMedCrossRef
4.
Zurück zum Zitat Beger HG, Rau B, Isenmann R, Schwarz M, Gansauge F, Poch B. Antibiotic prophylaxis in severe acute pancreatitis. Pancreatology 2005;5(1):10–19.PubMedCrossRef Beger HG, Rau B, Isenmann R, Schwarz M, Gansauge F, Poch B. Antibiotic prophylaxis in severe acute pancreatitis. Pancreatology 2005;5(1):10–19.PubMedCrossRef
5.
Zurück zum Zitat Beger HG, Bittner R, Block S, Buchler M. Bacterial contamination of pancreatic necrosis. A prospective clinical study. Gastroenterology 1986;91(2):433–438.PubMed Beger HG, Bittner R, Block S, Buchler M. Bacterial contamination of pancreatic necrosis. A prospective clinical study. Gastroenterology 1986;91(2):433–438.PubMed
6.
Zurück zum Zitat Van Felius ID, Akkermans LM, Bosscha K, Verheem A, Harmsen W, Visser MR, et al. Interdigestive small bowel motility and duodenal bacterial overgrowth in experimental acute pancreatitis. Neurogastroenterol Motil 2003;15(3):267–276.PubMedCrossRef Van Felius ID, Akkermans LM, Bosscha K, Verheem A, Harmsen W, Visser MR, et al. Interdigestive small bowel motility and duodenal bacterial overgrowth in experimental acute pancreatitis. Neurogastroenterol Motil 2003;15(3):267–276.PubMedCrossRef
7.
Zurück zum Zitat Berg RD. Bacterial translocation from the gastrointestinal tract. Adv Exp Med Biol 1999;473:11–30.PubMed Berg RD. Bacterial translocation from the gastrointestinal tract. Adv Exp Med Biol 1999;473:11–30.PubMed
8.
9.
Zurück zum Zitat Berg RD. Bacterial translocation from the gastrointestinal tract. Trends Microbiol 1995;3(4):149–154.PubMedCrossRef Berg RD. Bacterial translocation from the gastrointestinal tract. Trends Microbiol 1995;3(4):149–154.PubMedCrossRef
10.
Zurück zum Zitat Berg RD. Inhibition of bacterial translocation from the gastrointestinal tract to the mesenteric lymph nodes in specific pathogen-free mice but not gnotobiotic mice by non-specific macrophage activation. Adv Exp Med Biol 1995;371A:447–452.PubMed Berg RD. Inhibition of bacterial translocation from the gastrointestinal tract to the mesenteric lymph nodes in specific pathogen-free mice but not gnotobiotic mice by non-specific macrophage activation. Adv Exp Med Biol 1995;371A:447–452.PubMed
11.
Zurück zum Zitat Gautreaux MD, Deitch EA, Berg RD. T lymphocytes in host defense against bacterial translocation from the gastrointestinal tract. Infect Immun 1994;62(7):2874–2884.PubMed Gautreaux MD, Deitch EA, Berg RD. T lymphocytes in host defense against bacterial translocation from the gastrointestinal tract. Infect Immun 1994;62(7):2874–2884.PubMed
12.
Zurück zum Zitat Gianotti L, Munda R, Alexander JW, Tchervenkov JI, Babcock GF. Bacterial translocation: a potential source for infection in acute pancreatitis. Pancreas 1993;8(5):551–558.PubMedCrossRef Gianotti L, Munda R, Alexander JW, Tchervenkov JI, Babcock GF. Bacterial translocation: a potential source for infection in acute pancreatitis. Pancreas 1993;8(5):551–558.PubMedCrossRef
13.
Zurück zum Zitat Kazantsev GB, Hecht DW, Rao R, Fedorak IJ, Gattuso P, Thompson K et al. Plasmid labeling confirms bacterial translocation in pancreatitis. Am J Surg 1994;167(1):201–206.PubMedCrossRef Kazantsev GB, Hecht DW, Rao R, Fedorak IJ, Gattuso P, Thompson K et al. Plasmid labeling confirms bacterial translocation in pancreatitis. Am J Surg 1994;167(1):201–206.PubMedCrossRef
14.
Zurück zum Zitat Medich DS, Lee TK, Melhem MF, Rowe MI, Schraut WH, Lee KK. Pathogenesis of pancreatic sepsis. Am J Surg 1993;165(1):46–50.PubMedCrossRef Medich DS, Lee TK, Melhem MF, Rowe MI, Schraut WH, Lee KK. Pathogenesis of pancreatic sepsis. Am J Surg 1993;165(1):46–50.PubMedCrossRef
15.
Zurück zum Zitat Cicalese L, Sahai A, Sileri P, Rastellini C, Subbotin V, Ford H, et al. Acute pancreatitis and bacterial translocation. Dig Dis Sci 2001;46(5):1127–1132.PubMedCrossRef Cicalese L, Sahai A, Sileri P, Rastellini C, Subbotin V, Ford H, et al. Acute pancreatitis and bacterial translocation. Dig Dis Sci 2001;46(5):1127–1132.PubMedCrossRef
16.
Zurück zum Zitat Wells CL, Hess DJ, Erlandsen SL. Impact of the indigenous flora in animal models of shock and sepsis. Shock 2004;22(6):562–568.PubMedCrossRef Wells CL, Hess DJ, Erlandsen SL. Impact of the indigenous flora in animal models of shock and sepsis. Shock 2004;22(6):562–568.PubMedCrossRef
17.
Zurück zum Zitat McGarr SE, Ridlon JM, Hylemon PB. Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. J Clin Gastroenterol 2005;39(2):98–109.PubMed McGarr SE, Ridlon JM, Hylemon PB. Diet, anaerobic bacterial metabolism, and colon cancer: a review of the literature. J Clin Gastroenterol 2005;39(2):98–109.PubMed
18.
Zurück zum Zitat Finegold SM, Sutter VL, Sugihara PT, Elder HA, Lehmann SM, Phillips RL. Fecal microbial flora in Seventh Day Adventist populations and control subjects. Am J Clin Nutr 1977;30(11):1781–1792.PubMed Finegold SM, Sutter VL, Sugihara PT, Elder HA, Lehmann SM, Phillips RL. Fecal microbial flora in Seventh Day Adventist populations and control subjects. Am J Clin Nutr 1977;30(11):1781–1792.PubMed
19.
Zurück zum Zitat Gusttafsson BE, Fitsgerald RJ. Alteration in intestinal microbial flora of rats with tail cups to prevent coprophagy. Proc Soc Exp Biol Med 1960;104:319–322. Gusttafsson BE, Fitsgerald RJ. Alteration in intestinal microbial flora of rats with tail cups to prevent coprophagy. Proc Soc Exp Biol Med 1960;104:319–322.
20.
Zurück zum Zitat Jilge B, Meyer H. Coprophagy-dependant changes of the anaerobic bacterial flora in stomach and small intestine of the rabbit. Z Versuchstierkd 1975;17(5–6):308–314.PubMed Jilge B, Meyer H. Coprophagy-dependant changes of the anaerobic bacterial flora in stomach and small intestine of the rabbit. Z Versuchstierkd 1975;17(5–6):308–314.PubMed
21.
Zurück zum Zitat Bijlsma PB, Peeters RA, Groot JA, Dekker PR, Taminiau JA, Van Der MR. Differential in vivo and in vitro intestinal permeability to lactulose and mannitol in animals and humans: a hypothesis. Gastroenterology 1995;108(3):687–696.PubMedCrossRef Bijlsma PB, Peeters RA, Groot JA, Dekker PR, Taminiau JA, Van Der MR. Differential in vivo and in vitro intestinal permeability to lactulose and mannitol in animals and humans: a hypothesis. Gastroenterology 1995;108(3):687–696.PubMedCrossRef
22.
Zurück zum Zitat Van Minnen LP, Besselink MG, Bosscha K, Van Leeuwen MS, Schipper ME, Gooszen HG. Colonic involvement in acute pancreatitis. A retrospective study of 16 patients. Dig Surg 2004;21(1):33–38.PubMedCrossRef Van Minnen LP, Besselink MG, Bosscha K, Van Leeuwen MS, Schipper ME, Gooszen HG. Colonic involvement in acute pancreatitis. A retrospective study of 16 patients. Dig Surg 2004;21(1):33–38.PubMedCrossRef
23.
Zurück zum Zitat Arendt T, Wendt M, Olszewski M, Falkenhagen U, Stoffregen C, Folsch UR. Cerulein-induced acute pancreatitis in rats—does bacterial translocation occur via a transperitoneal pathway? Pancreas 1997;15(3):291–296.PubMedCrossRef Arendt T, Wendt M, Olszewski M, Falkenhagen U, Stoffregen C, Folsch UR. Cerulein-induced acute pancreatitis in rats—does bacterial translocation occur via a transperitoneal pathway? Pancreas 1997;15(3):291–296.PubMedCrossRef
24.
Zurück zum Zitat Nevalainen TJ, Seppa A. Acute pancreatitis caused by closed duodenal loop in the rat. Scand J Gastroenterol 1975;10(5):521–527.PubMed Nevalainen TJ, Seppa A. Acute pancreatitis caused by closed duodenal loop in the rat. Scand J Gastroenterol 1975;10(5):521–527.PubMed
25.
Zurück zum Zitat Dickson AP, Foulis AK, Imrie CW. Histology and bacteriology of closed duodenal loop models of experimental acute pancreatitis in the rat. Digestion 1986;34(1):15–21.PubMed Dickson AP, Foulis AK, Imrie CW. Histology and bacteriology of closed duodenal loop models of experimental acute pancreatitis in the rat. Digestion 1986;34(1):15–21.PubMed
26.
Zurück zum Zitat Habold C, Chevalier C, Dunel-Erb S, Foltzer-Jourdainne C, Le Maho Y, Lignot JH. Effects of fasting and refeeding on jejunal morphology and cellular activity in rats in relation to depletion of body stores. Scand J Gastroenterol 2004;39(6):531–539.PubMedCrossRef Habold C, Chevalier C, Dunel-Erb S, Foltzer-Jourdainne C, Le Maho Y, Lignot JH. Effects of fasting and refeeding on jejunal morphology and cellular activity in rats in relation to depletion of body stores. Scand J Gastroenterol 2004;39(6):531–539.PubMedCrossRef
27.
Zurück zum Zitat Deitch EA, Sittig K, Li M, Berg R, Specian RD. Obstructive jaundice promotes bacterial translocation from the gut. Am J Surg 1990;159(1):79–84.PubMedCrossRef Deitch EA, Sittig K, Li M, Berg R, Specian RD. Obstructive jaundice promotes bacterial translocation from the gut. Am J Surg 1990;159(1):79–84.PubMedCrossRef
28.
Zurück zum Zitat Nieuwenhuijs VB, Van Dijk JE, Gooszen HG, et al. Obstructive jaundice, bacterial translocation and indigestive small bowel motility in rats. Digestion 2000;62:255–261. Nieuwenhuijs VB, Van Dijk JE, Gooszen HG, et al. Obstructive jaundice, bacterial translocation and indigestive small bowel motility in rats. Digestion 2000;62:255–261.
29.
Zurück zum Zitat Ogata Y, Nishi M, Nakayama H, Kuwahara T, Ohnishi Y, Tashiro S. Role of bile in intestinal barrier function and its inhibitory effect on bacterial translocation in obstructive jaundice in rats. J Surg Res 2003;115(1):18–23.PubMedCrossRef Ogata Y, Nishi M, Nakayama H, Kuwahara T, Ohnishi Y, Tashiro S. Role of bile in intestinal barrier function and its inhibitory effect on bacterial translocation in obstructive jaundice in rats. J Surg Res 2003;115(1):18–23.PubMedCrossRef
30.
Zurück zum Zitat Lombardi B, Estes LW, Longnecker DS. Acute hemorrhagic pancreatitis (massive necrosis) with fat necrosis induced in mice by DL-ethionine fed with a choline-deficient diet. Am J Pathol 1975;79(3):465–480.PubMed Lombardi B, Estes LW, Longnecker DS. Acute hemorrhagic pancreatitis (massive necrosis) with fat necrosis induced in mice by DL-ethionine fed with a choline-deficient diet. Am J Pathol 1975;79(3):465–480.PubMed
31.
Zurück zum Zitat Lombardi B. Influence of dietary factors on the pancreatotoxicity of ethionine. Am J Pathol 1976;84(3):633–648.PubMed Lombardi B. Influence of dietary factors on the pancreatotoxicity of ethionine. Am J Pathol 1976;84(3):633–648.PubMed
32.
Zurück zum Zitat Niederau C, Luthen R, Niederau MC, Grendell JH, Ferrell LD. Acute experimental hemorrhagic-necrotizing pancreatitis induced by feeding a choline-deficient, ethionine-supplemented diet. Methodology and standards. Eur Surg Res 1992;24(Suppl 1):40–54.PubMed Niederau C, Luthen R, Niederau MC, Grendell JH, Ferrell LD. Acute experimental hemorrhagic-necrotizing pancreatitis induced by feeding a choline-deficient, ethionine-supplemented diet. Methodology and standards. Eur Surg Res 1992;24(Suppl 1):40–54.PubMed
33.
Zurück zum Zitat Rattner DW, Compton CC, Gu ZY, Wilkinson R, Warshaw AL. Bacterial infection is not necessary for lethal necrotizing pancreatitis in mice. Int J Pancreatol 1989;5(1):99–105.PubMed Rattner DW, Compton CC, Gu ZY, Wilkinson R, Warshaw AL. Bacterial infection is not necessary for lethal necrotizing pancreatitis in mice. Int J Pancreatol 1989;5(1):99–105.PubMed
34.
Zurück zum Zitat Churg A, Richter WR. Early changes in the exocrine pancreas of the dog and rat after ligation of the pancreatic duct. A light and electron microscopic study. Am J Pathol 1971;63(3):521–546.PubMed Churg A, Richter WR. Early changes in the exocrine pancreas of the dog and rat after ligation of the pancreatic duct. A light and electron microscopic study. Am J Pathol 1971;63(3):521–546.PubMed
35.
Zurück zum Zitat Edstrom C, Falkmer S. Pancreatic morphology and blood glucose level in rats at various intervals after duct ligation. Virchows Arch A Pathol Anat Histopathol 1968;345(2):139–153. Edstrom C, Falkmer S. Pancreatic morphology and blood glucose level in rats at various intervals after duct ligation. Virchows Arch A Pathol Anat Histopathol 1968;345(2):139–153.
36.
Zurück zum Zitat Lerch MM, Saluja AK, Runzi M, Dawra R, Saluja M, Steer ML. Pancreatic duct obstruction triggers acute necrotizing pancreatitis in the opossum. Gastroenterology 1993;104(3):853–861.PubMed Lerch MM, Saluja AK, Runzi M, Dawra R, Saluja M, Steer ML. Pancreatic duct obstruction triggers acute necrotizing pancreatitis in the opossum. Gastroenterology 1993;104(3):853–861.PubMed
37.
Zurück zum Zitat Lerch MM, Saluja AK, Runzi M, Dawra R, Steer ML. Luminal endocytosis and intracellular targeting by acinar cells during early biliary pancreatitis in the opossum. J Clin Invest 1995;95(5):2222–2231.PubMedCrossRef Lerch MM, Saluja AK, Runzi M, Dawra R, Steer ML. Luminal endocytosis and intracellular targeting by acinar cells during early biliary pancreatitis in the opossum. J Clin Invest 1995;95(5):2222–2231.PubMedCrossRef
38.
Zurück zum Zitat Cohen DB, Magnotti LJ, Lu Q, Xu dZ, Berezina TL, Zaets SB et al. Pancreatic duct ligation reduces lung injury following trauma and hemorrhagic shock. Ann Surg 2004;240(5):885–891.PubMedCrossRef Cohen DB, Magnotti LJ, Lu Q, Xu dZ, Berezina TL, Zaets SB et al. Pancreatic duct ligation reduces lung injury following trauma and hemorrhagic shock. Ann Surg 2004;240(5):885–891.PubMedCrossRef
39.
Zurück zum Zitat Deitch EA, Shi HP, Lu Q, Feketeova E, Xu dZ. Serine proteases are involved in the pathogenesis of trauma-hemorrhagic shock-induced gut and lung injury. Shock 2003;19(5):452–456.PubMedCrossRef Deitch EA, Shi HP, Lu Q, Feketeova E, Xu dZ. Serine proteases are involved in the pathogenesis of trauma-hemorrhagic shock-induced gut and lung injury. Shock 2003;19(5):452–456.PubMedCrossRef
40.
Zurück zum Zitat Sheen-Chen SM, Eng HL, Hung KS. Altered serum transforming growth factor-beta1 and monocyte chemoattractant protein-1 levels in obstructive jaundice. World J Surg 2004;28(10):967–970.PubMedCrossRef Sheen-Chen SM, Eng HL, Hung KS. Altered serum transforming growth factor-beta1 and monocyte chemoattractant protein-1 levels in obstructive jaundice. World J Surg 2004;28(10):967–970.PubMedCrossRef
41.
Zurück zum Zitat Parks RW, Halliday MI, McCrory DC, Erwin P, Smye M, Diamond T, et al. Host immune responses and intestinal permeability in patients with jaundice. Br J Surg 2003;90(2):239–245.PubMedCrossRef Parks RW, Halliday MI, McCrory DC, Erwin P, Smye M, Diamond T, et al. Host immune responses and intestinal permeability in patients with jaundice. Br J Surg 2003;90(2):239–245.PubMedCrossRef
42.
Zurück zum Zitat Nehez L, Andersson R. Compromise of immune function in obstructive jaundice. Eur J Surg 2002;168(6):315–328.PubMedCrossRef Nehez L, Andersson R. Compromise of immune function in obstructive jaundice. Eur J Surg 2002;168(6):315–328.PubMedCrossRef
43.
Zurück zum Zitat Lampel M, Kern HF. Acute interstitial pancreatitis in the rat induced by excessive doses of a pancreatic secretagogue. Virchows Arch A Pathol Anat Histopathol 1977;373(2):97–117.CrossRef Lampel M, Kern HF. Acute interstitial pancreatitis in the rat induced by excessive doses of a pancreatic secretagogue. Virchows Arch A Pathol Anat Histopathol 1977;373(2):97–117.CrossRef
44.
Zurück zum Zitat Niederau C, Ferrell LD, Grendell JH. Caerulein-induced acute necrotizing pancreatitis in mice: protective effects of proglumide, benzotript, and secretin. Gastroenterology 1985;88(5 Pt 1):1192–1204.PubMed Niederau C, Ferrell LD, Grendell JH. Caerulein-induced acute necrotizing pancreatitis in mice: protective effects of proglumide, benzotript, and secretin. Gastroenterology 1985;88(5 Pt 1):1192–1204.PubMed
45.
Zurück zum Zitat Willemer S, Elsasser HP, Adler G. Hormone-induced pancreatitis. Eur Surg Res 1992;24(Suppl 1):29–39.PubMed Willemer S, Elsasser HP, Adler G. Hormone-induced pancreatitis. Eur Surg Res 1992;24(Suppl 1):29–39.PubMed
46.
Zurück zum Zitat Novak D. Acceleration of small intestine contrast study by ceruletide. Gastrointest Radiol 1980;5(1):61–65.PubMedCrossRef Novak D. Acceleration of small intestine contrast study by ceruletide. Gastrointest Radiol 1980;5(1):61–65.PubMedCrossRef
47.
Zurück zum Zitat Runkel NS, Moody FG, Smith GS, Rodriguez LF, Larocco MT, Miller TA. The role of the gut in the development of sepsis in acute pancreatitis. J Surg Res 1991;51(1):18–23.PubMedCrossRef Runkel NS, Moody FG, Smith GS, Rodriguez LF, Larocco MT, Miller TA. The role of the gut in the development of sepsis in acute pancreatitis. J Surg Res 1991;51(1):18–23.PubMedCrossRef
48.
Zurück zum Zitat Armstrong CP, Taylor TV, Torrance HB. Effects of bile, infection and pressure on pancreatic duct integrity. Br J Surg 1985;72(10):792–795.PubMedCrossRef Armstrong CP, Taylor TV, Torrance HB. Effects of bile, infection and pressure on pancreatic duct integrity. Br J Surg 1985;72(10):792–795.PubMedCrossRef
49.
Zurück zum Zitat Armstrong CP, Taylor TV, Torrance HB. Pressure, volume and the pancreas. Gut 1985;26(6):615–624.PubMed Armstrong CP, Taylor TV, Torrance HB. Pressure, volume and the pancreas. Gut 1985;26(6):615–624.PubMed
50.
Zurück zum Zitat Lightner AM, Kirkwood KS. Pathophysiology of gallstone pancreatitis. Front Biosci 2001;6:E66–E76.PubMed Lightner AM, Kirkwood KS. Pathophysiology of gallstone pancreatitis. Front Biosci 2001;6:E66–E76.PubMed
51.
Zurück zum Zitat Aho HJ, Suonpaa K, Ahola RA, Nevalainen TJ. Experimental pancreatitis in the rat. Ductal factors in sodium taurocholate-induced acute pancreatitis. Exp Pathol 1984;25(2):73–79.PubMed Aho HJ, Suonpaa K, Ahola RA, Nevalainen TJ. Experimental pancreatitis in the rat. Ductal factors in sodium taurocholate-induced acute pancreatitis. Exp Pathol 1984;25(2):73–79.PubMed
52.
Zurück zum Zitat Schmidt J, Rattner DW, Lewandrowski K, Compton CC, Mandavilli U, Knoefel WT, et al. A better model of acute pancreatitis for evaluating therapy. Ann Surg 1992;215(1):44–56.PubMedCrossRef Schmidt J, Rattner DW, Lewandrowski K, Compton CC, Mandavilli U, Knoefel WT, et al. A better model of acute pancreatitis for evaluating therapy. Ann Surg 1992;215(1):44–56.PubMedCrossRef
53.
Zurück zum Zitat Foitzik T, Mithofer K, Ferraro MJ, Fernandez-del Castillo C, Lewandrowski KB, Rattner DW, Warshaw AL. Time course of bacterial infection of the pancreas and its relation to disease severity in a rodent model of acute necrotizing pancreatitis. Ann Surg 1994;220(2):193–198.PubMedCrossRef Foitzik T, Mithofer K, Ferraro MJ, Fernandez-del Castillo C, Lewandrowski KB, Rattner DW, Warshaw AL. Time course of bacterial infection of the pancreas and its relation to disease severity in a rodent model of acute necrotizing pancreatitis. Ann Surg 1994;220(2):193–198.PubMedCrossRef
54.
Zurück zum Zitat Beger HG, Rau B, Mayer J, Pralle U. Natural course of acute pancreatitis. World J Surg 1997;21(2):130–135.PubMedCrossRef Beger HG, Rau B, Mayer J, Pralle U. Natural course of acute pancreatitis. World J Surg 1997;21(2):130–135.PubMedCrossRef
55.
56.
Zurück zum Zitat Rattner DW. Experimental models of acute pancreatitis and their relevance to human disease. Scand J Gastroenterol Suppl 1996;219:6–9.PubMed Rattner DW. Experimental models of acute pancreatitis and their relevance to human disease. Scand J Gastroenterol Suppl 1996;219:6–9.PubMed
57.
Zurück zum Zitat Mithofer K, Fernandez-Del Castillo C, Ferraro MJ, Lewandrowski K, Rattner DW, Warshaw AL. Antibiotic treatment improves survival in experimental acute necrotizing pancreatitis. Gastroenterology 1996;110(1):232–240.PubMedCrossRef Mithofer K, Fernandez-Del Castillo C, Ferraro MJ, Lewandrowski K, Rattner DW, Warshaw AL. Antibiotic treatment improves survival in experimental acute necrotizing pancreatitis. Gastroenterology 1996;110(1):232–240.PubMedCrossRef
58.
Zurück zum Zitat Lankisch PG, Pohl U, Otto J, Rahlf G. When should treatment of acute experimental pancreatitis be started? The early phase of bile-induced acute pancreatitis. Res Exp Med (Berl) 1988;188(2):123–129.CrossRef Lankisch PG, Pohl U, Otto J, Rahlf G. When should treatment of acute experimental pancreatitis be started? The early phase of bile-induced acute pancreatitis. Res Exp Med (Berl) 1988;188(2):123–129.CrossRef
59.
Zurück zum Zitat Rattner DW. Experimental models of acute pancreatitis and their relevance to human disease. Scand J Gastroenterol Suppl 1996;219:6–9.PubMed Rattner DW. Experimental models of acute pancreatitis and their relevance to human disease. Scand J Gastroenterol Suppl 1996;219:6–9.PubMed
60.
Zurück zum Zitat Dickson AP, Foulis AK, Imrie CW. Histology and bacteriology of closed duodenal loop models of experimental acute pancreatitis in the rat. Digestion 1986;34(1):15–21.PubMedCrossRef Dickson AP, Foulis AK, Imrie CW. Histology and bacteriology of closed duodenal loop models of experimental acute pancreatitis in the rat. Digestion 1986;34(1):15–21.PubMedCrossRef
61.
Zurück zum Zitat Nieuwenhuijs VB, Verheem A, Duijvenbode-Beumer H, et al. The role of interdigestive small bowel motility in the regulation of gut microflora, bacterial overgrowth and bacterial translocation in rats, Ann Surg 1998;228:188–193.PubMedCrossRef Nieuwenhuijs VB, Verheem A, Duijvenbode-Beumer H, et al. The role of interdigestive small bowel motility in the regulation of gut microflora, bacterial overgrowth and bacterial translocation in rats, Ann Surg 1998;228:188–193.PubMedCrossRef
62.
Zurück zum Zitat Runkel NS, Smith GS, Rodriguez LF, Larocco MT, Moody FG, Miller TA. Influence of shock on development of infection during acute pancreatitis in the rat. Dig Dis Sci 1992;37(9):1418–1425.PubMedCrossRef Runkel NS, Smith GS, Rodriguez LF, Larocco MT, Moody FG, Miller TA. Influence of shock on development of infection during acute pancreatitis in the rat. Dig Dis Sci 1992;37(9):1418–1425.PubMedCrossRef
63.
Zurück zum Zitat Velin AK, Ericson AC, Braaf Y, Wallon C, Soderholm JD. Increased antigen and bacterial uptake in follicle associated epithelium induced by chronic psychological stress in rats. Gut 2004;53(4):494–500.PubMedCrossRef Velin AK, Ericson AC, Braaf Y, Wallon C, Soderholm JD. Increased antigen and bacterial uptake in follicle associated epithelium induced by chronic psychological stress in rats. Gut 2004;53(4):494–500.PubMedCrossRef
64.
Zurück zum Zitat Moody FG, Haley-Russell D, Muncy DM. Intestinal transit and bacterial translocation in obstructive pancreatitis. Dig Dis Sci 1995;40(8):1798–1804.PubMedCrossRef Moody FG, Haley-Russell D, Muncy DM. Intestinal transit and bacterial translocation in obstructive pancreatitis. Dig Dis Sci 1995;40(8):1798–1804.PubMedCrossRef
65.
Zurück zum Zitat Liu Q, Djuricin G, Rossi H, Bewsey K, Nathan C, Gattuso P, et al. The effect of lexipafant on bacterial translocation in acute necrotizing pancreatitis in rats. Am Surg 1999;65(7):611–616.PubMed Liu Q, Djuricin G, Rossi H, Bewsey K, Nathan C, Gattuso P, et al. The effect of lexipafant on bacterial translocation in acute necrotizing pancreatitis in rats. Am Surg 1999;65(7):611–616.PubMed
66.
Zurück zum Zitat de Souza LJ, Sampietre SN, Assis RS, Knowles CH, Leite KR, Jancar S et al. Effect of platelet-activating factor antagonists (BN-52021, WEB-2170, and BB-882) on bacterial translocation in acute pancreatitis. J Gastrointest Surg 2001;5(4):364–370.PubMedCrossRef de Souza LJ, Sampietre SN, Assis RS, Knowles CH, Leite KR, Jancar S et al. Effect of platelet-activating factor antagonists (BN-52021, WEB-2170, and BB-882) on bacterial translocation in acute pancreatitis. J Gastrointest Surg 2001;5(4):364–370.PubMedCrossRef
67.
Zurück zum Zitat Inoue K, Hirota M, Kimura Y, Kuwata K, Ohmuraya M, Ogawa M. Further evidence for endothelin as an important mediator of pancreatic and intestinal ischemia in severe acute pancreatitis. Pancreas 2003;26(3):218–223.PubMedCrossRef Inoue K, Hirota M, Kimura Y, Kuwata K, Ohmuraya M, Ogawa M. Further evidence for endothelin as an important mediator of pancreatic and intestinal ischemia in severe acute pancreatitis. Pancreas 2003;26(3):218–223.PubMedCrossRef
68.
Zurück zum Zitat Yasuda T, Takeyama Y, Ueda T, Hori Y, Nishikawa J, Kuroda Y. Nonocclusive visceral ischemia associated with severe acute pancreatitis. Pancreas 2003;26(1):95–97.PubMedCrossRef Yasuda T, Takeyama Y, Ueda T, Hori Y, Nishikawa J, Kuroda Y. Nonocclusive visceral ischemia associated with severe acute pancreatitis. Pancreas 2003;26(1):95–97.PubMedCrossRef
69.
Zurück zum Zitat Rahman SH, Ammori BJ, Holmfield J, Larvin M, McMahon MJ. Intestinal hypoperfusion contributes to gut barrier failure in severe acute pancreatitis. J Gastrointest Surg 2003;7(1):26–35.PubMedCrossRef Rahman SH, Ammori BJ, Holmfield J, Larvin M, McMahon MJ. Intestinal hypoperfusion contributes to gut barrier failure in severe acute pancreatitis. J Gastrointest Surg 2003;7(1):26–35.PubMedCrossRef
70.
Zurück zum Zitat McKay CJ, Imrie CW. The continuing challenge of early mortality in acute pancreatitis. Br J Surg 2004;91(10):1243–1244.PubMedCrossRef McKay CJ, Imrie CW. The continuing challenge of early mortality in acute pancreatitis. Br J Surg 2004;91(10):1243–1244.PubMedCrossRef
71.
Zurück zum Zitat Tran DD, Cuesta MA, Schneider AJ, Wesdorp RI. Prevalence and prediction of multiple organ system failure and mortality in acute pancreatitis. J Crit Care 1993;8(3):145–153.PubMedCrossRef Tran DD, Cuesta MA, Schneider AJ, Wesdorp RI. Prevalence and prediction of multiple organ system failure and mortality in acute pancreatitis. J Crit Care 1993;8(3):145–153.PubMedCrossRef
72.
Zurück zum Zitat Bhatia M, Wong FL, Cao Y, Lau HY, Huang J, Puneet P, et al. Pathophysiology of acute pancreatitis. Pancreatology 2005;5(2–3):132–144.PubMedCrossRef Bhatia M, Wong FL, Cao Y, Lau HY, Huang J, Puneet P, et al. Pathophysiology of acute pancreatitis. Pancreatology 2005;5(2–3):132–144.PubMedCrossRef
73.
Zurück zum Zitat Bhatia M. Inflammatory response on the pancreatic acinar cell injury. Scand J Surg 2005;94(2):97–102.PubMed Bhatia M. Inflammatory response on the pancreatic acinar cell injury. Scand J Surg 2005;94(2):97–102.PubMed
74.
Zurück zum Zitat Gloor B, Muller CA, Worni M, Martignoni ME, Uhl W, Buchler MW. Late mortality in patients with severe acute pancreatitis. Br J Surg 2001;88(7):975–979.PubMedCrossRef Gloor B, Muller CA, Worni M, Martignoni ME, Uhl W, Buchler MW. Late mortality in patients with severe acute pancreatitis. Br J Surg 2001;88(7):975–979.PubMedCrossRef
75.
Zurück zum Zitat Dugernier T, Laterre PF, Reynaert M, Deby-Dupont G. Compartmentalization of the protease–antiprotease balance in early severe acute pancreatitis. Pancreas 2005;31(2):168–173.PubMedCrossRef Dugernier T, Laterre PF, Reynaert M, Deby-Dupont G. Compartmentalization of the protease–antiprotease balance in early severe acute pancreatitis. Pancreas 2005;31(2):168–173.PubMedCrossRef
76.
Zurück zum Zitat Wilson PG, Manji M, Neoptolemos JP. Acute pancreatitis as a model of sepsis. J Antimicrob Chemother 1998;41(Suppl A):51–63.PubMedCrossRef Wilson PG, Manji M, Neoptolemos JP. Acute pancreatitis as a model of sepsis. J Antimicrob Chemother 1998;41(Suppl A):51–63.PubMedCrossRef
77.
Zurück zum Zitat Webster MW, Pasculle AW, Myerowitz RL, Rao KN, Lombardi B. Postinduction bacteremia in experimental acute pancreatitis. Am J Surg 1979;138(3):418–420.PubMedCrossRef Webster MW, Pasculle AW, Myerowitz RL, Rao KN, Lombardi B. Postinduction bacteremia in experimental acute pancreatitis. Am J Surg 1979;138(3):418–420.PubMedCrossRef
78.
Zurück zum Zitat Redan JA, Rush BF, McCullough JN, Machiedo GW, Murphy TF, Dikdan GS, et al. Organ distribution of radiolabeled enteric Escherichia coli during and after hemorrhagic shock. Ann Surg 1990;211(6):663–666.PubMedCrossRef Redan JA, Rush BF, McCullough JN, Machiedo GW, Murphy TF, Dikdan GS, et al. Organ distribution of radiolabeled enteric Escherichia coli during and after hemorrhagic shock. Ann Surg 1990;211(6):663–666.PubMedCrossRef
79.
Zurück zum Zitat Runkel NS, Rodriguez LF, Moody FG. Mechanisms of sepsis in acute pancreatitis in opossums. Am J Surg 1995;169(2):227–232.PubMedCrossRef Runkel NS, Rodriguez LF, Moody FG. Mechanisms of sepsis in acute pancreatitis in opossums. Am J Surg 1995;169(2):227–232.PubMedCrossRef
80.
Zurück zum Zitat Widdison AL, Karanjia ND, Reber HA. Routes of spread of pathogens into the pancreas in a feline model of acute pancreatitis. Gut 1994;35(9):1306–1310.PubMed Widdison AL, Karanjia ND, Reber HA. Routes of spread of pathogens into the pancreas in a feline model of acute pancreatitis. Gut 1994;35(9):1306–1310.PubMed
81.
Zurück zum Zitat Samel S, Lanig S, Lux A, Keese M, Gretz N, Nichterlein T, et al. The gut origin of bacterial pancreatic infection during acute experimental pancreatitis in rats. Pancreatology 2002;2(5):449–455.PubMedCrossRef Samel S, Lanig S, Lux A, Keese M, Gretz N, Nichterlein T, et al. The gut origin of bacterial pancreatic infection during acute experimental pancreatitis in rats. Pancreatology 2002;2(5):449–455.PubMedCrossRef
Metadaten
Titel
The Use of Animal Models to Study Bacterial Translocation During Acute Pancreatitis
verfasst von
L. P. van Minnen
M. Blom
H. M. Timmerman
M. R. Visser
H. G. Gooszen
L. M. A. Akkermans
Publikationsdatum
01.05.2007
Verlag
Springer-Verlag
Erschienen in
Journal of Gastrointestinal Surgery / Ausgabe 5/2007
Print ISSN: 1091-255X
Elektronische ISSN: 1873-4626
DOI
https://doi.org/10.1007/s11605-007-0088-0

Weitere Artikel der Ausgabe 5/2007

Journal of Gastrointestinal Surgery 5/2007 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.